201
|
Yano S, Ashida K, Nagata H, Ohe K, Wada N, Takeichi Y, Hanada Y, Ibayashi Y, Wang L, Sakamoto S, Sakamoto R, Uchi H, Shiratsuchi M, Furue M, Nomura M, Ogawa Y. Nivolumab-induced thyroid dysfunction lacking antithyroid antibody is frequently evoked in Japanese patients with malignant melanoma. BMC Endocr Disord 2018; 18:36. [PMID: 29884162 PMCID: PMC5994101 DOI: 10.1186/s12902-018-0267-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 06/01/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Nivolumab, an anti-programmed cell death-1 monoclonal antibody, has improved the survival of patients with malignant melanoma. Despite its efficacy, nivolumab inconsistently induces thyroid dysfunction as an immune-related adverse event (irAE). This study aimed to evaluate nivolumab-induced thyroid dysfunction to determine the risks and mechanisms of thyroid irAEs. METHODS After excluding 10 patients, data of 24 patients with malignant melanoma (aged 17-85 years; 54% female) were retrospectively analyzed. RESULTS Thyroid irAEs were observed in seven patients (29%). Three patients had hypothyroidism after preceding transient thyrotoxicosis, and the other four patients had hypothyroidism without thyrotoxicosis. Levothyroxine-Na replacement was required in three patients. Antithyroid antibody (ATA) titer was elevated in one of four assessable patients. The average (±SD) time to onset of thyroid irAE was 33.6 (±21.9) weeks. The administration period of nivolumab was longer in patients with thyroid irAEs than in those without thyroid irAEs (P < 0.01). There were no significant differences between patients with and without thyroid irAEs regarding age, sex, tumor stage, response to nivolumab therapy, baseline thyroid function, antithyroid peroxidase antibody (anti-TPO Ab) and antithyroglobulin antibody (anti-Tg Ab). CONCLUSIONS Thyroid dysfunction was a common irAE of nivolumab in malignant melanoma. Neither anti-TPO Ab nor anti-Tg Ab was associated with the risk for nivolumab-induced thyroid dysfunction. A conventional ATA-independent mechanism might be involved in thyroid irAEs. Further studies are required to clarify the mechanism and identify the predictive factors of thyroid irAEs.
Collapse
Affiliation(s)
- Seiichi Yano
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Kenji Ashida
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Hiromi Nagata
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Kenji Ohe
- Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Naoko Wada
- Department of Dermatology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yukina Takeichi
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Yuki Hanada
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Yuta Ibayashi
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Lixiang Wang
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Shohei Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Hiroshi Uchi
- Department of Dermatology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Motoaki Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| | - Masutaka Furue
- Department of Dermatology, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011 Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School Of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582 Japan
| |
Collapse
|
202
|
De Sousa SMC, Sheriff N, Tran CH, Menzies AM, Tsang VHM, Long GV, Tonks KTT. Fall in thyroid stimulating hormone (TSH) may be an early marker of ipilimumab-induced hypophysitis. Pituitary 2018; 21:274-282. [PMID: 29380110 DOI: 10.1007/s11102-018-0866-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Hypophysitis develops in up to 19% of melanoma patients treated with ipilimumab, a cytotoxic T-lymphocyte antigen-4 antibody. Early detection may avert life-threatening hypopituitarism. We aimed to assess the incidence of ipilimumab-induced hypophysitis (IH) at a quaternary melanoma referral centre, and to determine whether cortisol or thyroid stimulating hormone (TSH) monitoring could predict IH onset. METHODS We performed a retrospective cohort study of ipilimumab-treated patients at a quaternary melanoma referral centre in Australia. The inclusion criteria were patients with metastatic or unresectable melanoma treated with ipilimumab monotherapy, and cortisol and TSH measurements prior to ≥ 2 infusions. The main outcomes were IH incidence and TSH and cortisol patterns in patients who did and did not develop IH. RESULTS Of 78 ipilimumab-treated patients, 46 met the study criteria and 9/46 (20%) developed IH at a median duration of 13.0 weeks (range 7.7-18.1) following ipilimumab initiation. All patients whose TSH fell ≥ 80% compared to baseline developed IH, and, in 5/9 patients with IH, TSH fell prior to cortisol fall and IH diagnosis. Pre-cycle-4 TSH was significantly lower in those who developed IH (0.31 vs. 1.73 mIU/L, P = 0.006). TSH fall was detected at a median time of 9.2 (range 7.7-16.4) weeks after commencing ipilimumab, and a median of 3.6 (range of - 1.4 to 9.7) weeks before IH diagnosis. There was no difference in TSH between the groups before cycles 1-3 or in cortisol before cycles 1-4. CONCLUSIONS TSH fall ≥ 80% may be an early marker of IH. Serial TSH measurement during ipilimumab therapy may be an inexpensive tool to expedite IH diagnosis.
Collapse
Affiliation(s)
- Sunita M C De Sousa
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, an SA Pathology and University of South Australia Alliance, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
| | - Nisa Sheriff
- Department of Endocrinology, St Vincent's Hospital, Darlinghurst, Australia
- Diabetes and Metabolism Program, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Chau H Tran
- Department of Endocrinology, St Vincent's Hospital, Darlinghurst, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, Wollstonecraft, Australia
- Royal North Shore Hospital, St Leonards, Australia
- University of Sydney, Camperdown, Australia
- Mater Hospital, North Sydney, Australia
| | - Venessa H M Tsang
- Royal North Shore Hospital, St Leonards, Australia
- Mater Hospital, North Sydney, Australia
- Kolling Institute of Medical Research, St Leonards, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Wollstonecraft, Australia
- Royal North Shore Hospital, St Leonards, Australia
- University of Sydney, Camperdown, Australia
- Mater Hospital, North Sydney, Australia
| | - Katherine T T Tonks
- Department of Endocrinology, St Vincent's Hospital, Darlinghurst, Australia.
- Diabetes and Metabolism Program, Garvan Institute of Medical Research, Darlinghurst, Australia.
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia.
- Mater Hospital, North Sydney, Australia.
| |
Collapse
|
203
|
Immunotherapy in Non-Small Cell Lung Cancer Treatment: Current Status and the Role of Imaging. J Thorac Imaging 2018; 32:300-312. [PMID: 28786858 DOI: 10.1097/rti.0000000000000291] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lung cancer remains the leading cause of cancer-related mortality and is responsible for more deaths than breast, prostate, and colon cancer combined. Most patients are diagnosed with advanced disease at the time of presentation, and treatment options have traditionally included surgery, chemotherapy, and/or radiation. However, significant advances in the molecular characterization of lung cancer have led to the creation of effective immunotherapies that assist in the recognition of cancer as foreign by the host immune system, stimulate the immune system, and relieve the inhibition that allows tumor growth and spread. Extensive experience with the immunomodulatory monoclonal antibody ipilimumab has demonstrated that unique responses may be seen with immunotherapies that are not adequately captured by traditional response criteria such as the World Health Organization criteria and Response Evaluation Criteria in Solid Tumors (RECIST). Consequently, several modified criteria have been developed to evaluate patients treated with immunotherapy, including immune-related response criteria, immune-related RECIST, and immune RECIST. Finally, patients undergoing immunotherapy may develop a wide variety of immune-related adverse events with which the radiologist must be familiar. In this article, we present the fundamental concepts behind immunotherapy, specific agents currently approved for the treatment of lung cancer, and immune-related adverse events. The role of imaging in the evaluation of these patients will also be discussed, including the general principles of treatment response evaluation, specific response criteria adopted with these agents, including immune-related response criteria, immune-related RECIST, and immune RECIST, and the imaging of immune-related adverse events.
Collapse
|
204
|
Lyu C, Li W, Liu S, Gao S, Zhang H, Hao L, Yu H, Wei W, Song J, Yang Y, Wang C, Zhang Z, Wang N. Systematic review on the efficacy and safety of immune checkpoint inhibition in renal cell carcinoma. Future Oncol 2018; 14:2207-2221. [PMID: 29726696 DOI: 10.2217/fon-2018-0193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To perform a systematic review of the relevant literature about clinical trials on efficacy and safety of immune checkpoint inhibition, whether it is used alone, in combination or with other targeted therapies in patients with advanced and metastatic renal cell carcinoma (RCC), two team members reviewed the abstracts and selected pertinent articles from the relevant databases. A narrative review of randomized controlled trials was performed and seven randomized controlled trials were identified in this systematic review. In treatment of RCC, nivolumab has superior efficacy and safety compared with second-line everolimus. Combination strategies, especially those combined with anti-VEGF agents presents better efficacy but worse outcomes in term of safety than monotherapy and conventional treatment and might guide treatment choice for patients with RCC.
Collapse
Affiliation(s)
- Chen Lyu
- College of Animal Science, Jilin University, Changchun, PR China
| | - Wenyue Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Songcai Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China.,Five-Star Animal Health Pharmaceutical Factory of Jilin Province, Changchun, PR China
| | - Shuang Gao
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Hong Zhang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, PR China
| | - Hao Yu
- College of Animal Science, Jilin University, Changchun, PR China
| | - Wenzhen Wei
- College of Animal Science, Jilin University, Changchun, PR China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, PR China
| | - Yaoyao Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Chunmeng Wang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Zhimin Zhang
- College of Animal Science, Jilin University, Changchun, PR China
| | - Nan Wang
- China Institute of Veterinary Drug Control, Beijing, PR China
| |
Collapse
|
205
|
Knochelmann HM, Dwyer CJ, Bailey SR, Amaya SM, Elston DM, Mazza-McCrann JM, Paulos CM. When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell Mol Immunol 2018; 15:458-469. [PMID: 29563615 PMCID: PMC6068176 DOI: 10.1038/s41423-018-0004-4] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
The balance between Th17 cells and regulatory T cells (Tregs) has emerged as a prominent factor in regulating autoimmunity and cancer. Th17 cells are vital for host defense against pathogens but have also been implicated in causing autoimmune disorders and cancer, though their role in carcinogenesis is less well understood. Tregs are required for self-tolerance and defense against autoimmunity and often correlate with cancer progression. This review addresses the importance of a functional homeostasis between these two subsets in health and the consequences of its disruption when these forces collide in disease. Importantly, we discuss the ability of Th17 cells to mediate cancer regression in immunotherapy, including adoptive transfer and checkpoint blockade therapy, and the therapeutic possibilities of purposefully offsetting the Th17/Treg balance to treat patients with cancer as well as those with autoimmune diseases.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA.
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Stefanie R Bailey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Sierra M Amaya
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Dirk M Elston
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Joni M Mazza-McCrann
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
206
|
Guaraldi F, La Selva R, Samà MT, D'Angelo V, Gori D, Fava P, Fierro MT, Savoia P, Arvat E. Characterization and implications of thyroid dysfunction induced by immune checkpoint inhibitors in real-life clinical practice: a long-term prospective study from a referral institution. J Endocrinol Invest 2018; 41:549-556. [PMID: 29043574 DOI: 10.1007/s40618-017-0772-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023]
Abstract
PURPOSE Autoimmune diseases are typically associated with immune checkpoints blockade. This study aims at assessing, in real-life clinical practice, the prevalence and impact of thyroid disorders induced by immune checkpoint inhibitors. METHODS 52 patients (30 F; age 61 ± 13 years) with advanced melanoma treated with ipilimumab (3 mg/kg i.v./3 weeks; 4 doses) were included. For disease progression, 29 (16 F) of them received nivolumab (3 mg/kg i.v./2 weeks) or pembrolizumab (2 mg/kg i.v./3 weeks). Thyroid function and autoimmunity were assessed before, after 6 weeks, at the end of ipilimumab, as well as before and every 3 months during nivolumab/pembrolizumab treatment. RESULTS During ipilimumab, 7 (4 F) patients developed thyroid dysfunction (4 thyroiditis, 1 associated with hypothyroidism; 2 thyrotoxicosis in a previously euthyroid multinodular goiter; 1 hypothyroidism worsened). During PD1 inhibitors, 7 patients (3 F) developed hypothyroidism with severe manifestations in 6 of them; 3 patients suffered from euthyroid autoimmune thyroiditis from baseline, one after ipilimumab; 2 patients developed after transient thyrotoxicosis. Mean follow-up after anti-CTLA4 inhibitors treatment was 36 ± 28 months. Thyroid disorders occurred 45.1 ± 20.8 and 151 ± 67 days after the initiation of CTLA4 and PD1 inhibitors, respectively. Autoimmune disorders and BRAF mutation were associated with a better clinical response to CTLA4 followed by PD1 treatment. CONCLUSIONS Immune checkpoint blockade is burdened by a high incidence of autoimmune thyroid dysfunction, which is often severe. Therefore, early and careful monitoring and, eventually, treatment are crucial to prevent the negative impact of thyroid dysfunction on the clinical outcome.
Collapse
Affiliation(s)
- F Guaraldi
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy.
- Pituitary Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), IRCCS Institute of Neurological Sciences of Bologna, University of Bologna, Via Altura 3, 40139, Bologna, Italy.
| | - R La Selva
- Division of Dermatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - M T Samà
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - V D'Angelo
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - D Gori
- Hygiene, Public Health and Medical Statistics Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - P Fava
- Pituitary Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), IRCCS Institute of Neurological Sciences of Bologna, University of Bologna, Via Altura 3, 40139, Bologna, Italy
| | - M T Fierro
- Pituitary Unit, Department of Biomedical and Neuromotor Sciences (DIBINEM), IRCCS Institute of Neurological Sciences of Bologna, University of Bologna, Via Altura 3, 40139, Bologna, Italy
| | - P Savoia
- Department of Health Sciences, "A. Avogadro" University of Eastern Piedmont, Novara, Italy
| | - E Arvat
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
207
|
Kähler KC, Eigentler TK, Gesierich A, Heinzerling L, Loquai C, Meier F, Meiss F, Pföhler C, Schlaak M, Terheyden P, Thoms KM, Ziemer M, Zimmer L, Gutzmer R. Ipilimumab in metastatic melanoma patients with pre-existing autoimmune disorders. Cancer Immunol Immunother 2018; 67:825-834. [PMID: 29487980 PMCID: PMC11028108 DOI: 10.1007/s00262-018-2134-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/16/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Ipilimumab and programmed death (PD) 1-antibodies are effective treatment options in metastatic melanoma. The safety and efficacy of ipilimumab in patients with pre-existing autoimmune disorders (AD) has only been evaluated in a selected number of patients. METHODS We performed a retrospective analysis in 14 German skin cancer centers for patients with metastatic melanoma and pre-existing AD treated with ipilimumab. RESULTS 41 patients with 44 pre-existing AD were treated with ipilimumab (thyroiditis n = 15, rheumatoid n = 11, dermatologic n = 10, Crohn's disease/ulcerative colitis n = 3, neurological n = 2, sarcoidosis n = 2, pancreatitis n = 1). 3 out of 41 patients had two AD, 11 patients required immunosuppressants at the time of induction of ipilimumab. 12 patients (29.2%) experienced a flare of their pre-existing AD, mainly patients with rheumatoid or dermatologic diseases. Additional immune-related adverse events (irAEs) occurred in 12 patients (29.2%). In 23 patients (56%) neither a change of their AD nor additional irAEs were observed. Objective responses were seen in five patients (one complete remission, four partial remissions, 12.1%). CONCLUSION This is the largest series of patients with pre-existing AD and treatment with ipilimumab reported. Flares of pre-existing AD were observed but manageable. Response rates and occurrence of new irAEs were comparable to previous trials. Thus, in this patient subgroup, ipilimumab can be a treatment option after a thorough discussion of pros and cons and taking severity and activity of the preexisting AD into account.
Collapse
Affiliation(s)
- Katharina C Kähler
- Department of Dermatology, Campus Kiel, University Hospital Schleswig-Holstein (UKSH), Rosalind-Franklind-Str. 7, 24105, Kiel, Germany.
| | - Thomas K Eigentler
- Department of Dermatology, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center of Mainz, Mainz, Germany
| | - Friedegund Meier
- Department of Dermatology, University of Dresden, Dresden, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/saar, Germany
| | - Max Schlaak
- Department of Dermatology, Skin Cancer Center at Center of Integrated Oncology (CIO), University Hospital Cologne, Cologne-Bonn, Germany
| | - Patrick Terheyden
- Department of Dermatology, Campus LübeckUniversity, Hospital (UKSH), Lübeck, Germany
| | - Kai M Thoms
- Department of Dermatology, University Medical Center Göttingen, Göttingen, Germany
| | - Mirjana Ziemer
- Department of Dermatology, University Hospital Leipzig, Leipzig, Germany
| | - Lisa Zimmer
- Department of Dermatology, Essen, Germany and German Cancer Consortium (DKTK), University Duisburg-Essen, University Hospital Essen, Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hanover, Germany
| |
Collapse
|
208
|
Gastrointestinal toxicity of immune checkpoint inhibitors: from mechanisms to management. Nat Rev Gastroenterol Hepatol 2018; 15:222-234. [PMID: 29512649 DOI: 10.1038/nrgastro.2018.14] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitor therapies are a novel group of monoclonal antibodies with proven effectiveness in a wide range of malignancies, including melanoma, renal cell carcinoma, non-small-cell lung cancer, urothelial carcinoma and Hodgkin lymphoma. Their use in a range of other indications, such as gastrointestinal and head and neck cancer, is currently under investigation. The number of agents included in this drug group is increasing, as is their use. Although they have the potential to improve the treatment of advanced malignancies, they are also associated with a substantial risk of immune-related adverse events. The incidence of gastrointestinal toxicity associated with their use is second only in frequency to dermatological toxicity. Thus, gastroenterologists can expect to be increasingly frequently consulted by oncologists as part of a multidisciplinary approach to managing toxicity. Here, we describe this novel group of agents and their mechanisms of action. We review the manifestations of gastrointestinal toxicity associated with their use so that it can be recognized early and diagnosed accurately. We also discuss the proposed mechanisms underlying this toxicity and describe an algorithmic and, wherever possible, evidence-based approach to its management.
Collapse
|
209
|
Sosa A, Lopez Cadena E, Simon Olive C, Karachaliou N, Rosell R. Clinical assessment of immune-related adverse events. Ther Adv Med Oncol 2018; 10:1758835918764628. [PMID: 29623110 PMCID: PMC5882039 DOI: 10.1177/1758835918764628] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy through checkpoint inhibitors is now standard practice for a growing number of cancer types, supported by overall improvement of clinical outcomes and better tolerance. One anti-CTLA-4 antibody (ipilimumab), two anti-PD-1 antibodies (pembrolizumab and nivolumab) and three anti-PD-L1 antibodies (atezolizumab, avelumab and durvalumab) have been approved for clear benefits across diverse trials. Adverse events of an immune nature associated with these agents frequently affect the skin, colon, endocrine glands, lungs and liver. Most of these effects are mild and can be managed through transient immunosuppression with corticosteroids, but high-grade events often require hospitalization and specialized treatment. However, since immunotherapy is recent, physicians with clinical experience in the diagnosis and management of immune toxicities are frequently those who actively participated in trials, but many practicing oncologists are still not familiarized with the assessment of these events. This review focuses on the incidence, diagnostic assessment and recommended management of the most relevant immune-related adverse events.
Collapse
Affiliation(s)
- Aaron Sosa
- Department of Medical Oncology, Hospital Universitari Sagrat Cor, Viladomat 288, Barcelona 08029, Spain
| | - Esther Lopez Cadena
- Department of Pneumonology, Hospital Universitari Sagrat Cor, Barcelona, Spain
| | | | - Niki Karachaliou
- Department of Medical Oncology, Hospital Universitari Sagrat Cor, Barcelona, Spain
| | - Rafael Rosell
- Department of Medical Oncology, Dr Rosell Oncology Institute, Barcelona, Spain
| |
Collapse
|
210
|
Eshghi N, Garland LL, Nia E, Betancourt R, Krupinski E, Kuo PH. 18F-FDG PET/CT Can Predict Development of Thyroiditis Due to Immunotherapy for Lung Cancer. J Nucl Med Technol 2018; 46:260-264. [PMID: 29599403 DOI: 10.2967/jnmt.117.204933] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022] Open
Abstract
Our primary purpose was to determine whether increased 18F-FDG uptake in the thyroid gland predicts development of thyroiditis with subsequent hypothyroidism in patients undergoing immunotherapy with nivolumab for lung cancer. Secondarily, we determined whether 18F-FDG uptake in the thyroid gland correlates with number of administered cycles of nivolumab. Methods: Retrospective chart review over 2 y found 18 lung cancer patients treated with nivolumab who underwent 18F-FDG PET/CT before and during therapy. SUVmean, SUVmax, and total lesion glycolysis of the thyroid gland were measured. SUVs were also measured for the pituitary gland, liver, and spleen. Patients underwent monthly thyroid testing. PET/CT parameters were analyzed by unpaired t testing for differences between 2 groups (patients who developed hypothyroidism and those who did not). Correlation between development of thyroiditis and number of cycles of nivolumab was also tested. Results: Six of 18 patients developed hypothyroidism. The t test comparing the 2 groups demonstrated significant differences in SUVmean (P = 0.04), SUVmax (P = 0.04), and total lesion glycolysis (P = 0.02) of the thyroid gland. Two of 4 patients who developed thyroiditis and had increased 18F-FDG uptake in the thyroid gland had a normal TSH level at the time of follow-up 18F-FDG PET/CT. Patients who developed thyroiditis with subsequent hypothyroidism stayed longer on therapy (10.6 cycles) than patients without thyroiditis (7.6 cycles), but the trend was not statistically significant. No significant difference in PET/CT parameters was observed for pituitary gland, liver, or spleen. Conclusion:18F-FDG PET/CT can predict the development of thyroiditis with subsequent hypothyroidism before laboratory testing. Further study is required to confirm the positive trend between thyroiditis and duration of therapy.
Collapse
Affiliation(s)
| | - Linda L Garland
- Department of Medicine, Section of Hematology and Medical Oncology, Banner University Medical Center, Tucson, Arizona
| | - Emily Nia
- Breast Imaging Section, Department of Radiology, University of Texas M.D. Anderson Cancer Center, Housten, Texas
| | - Robert Betancourt
- Department of Medicine, Banner University Medical Center, Tucson, Arizona
| | - Elizabeth Krupinski
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia; and
| | - Phillip H Kuo
- Departments of Medical Imaging and Medicine, Banner University Medical Center, and Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| |
Collapse
|
211
|
Kähler KC, Hassel JC, Heinzerling L, Loquai C, Mössner R, Ugurel S, Zimmer L, Gutzmer R. Management of side effects of immune checkpoint blockade by anti-CTLA-4 and anti-PD-1 antibodies in metastatic melanoma. J Dtsch Dermatol Ges 2018; 14:662-81. [PMID: 27373241 DOI: 10.1111/ddg.13047] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CTLA-4 and PD-1 are potential targets for tumor-induced downregulation of lymphocytic immune responses. Immune checkpoint-modifying monoclonal antibodies oppose these effects, inducing T cell-mediated immune responses to various tumors including melanoma. Both anti-CTLA-4 and anti-PD-1 antibodies modify the interaction between tumor, antigen-presenting cells, and T lymphocytes. With respect to overall survival, clinical studies have shown a major benefit for the anti-CTLA-4 antibody ipilimumab as well as the two anti-PD-1 antibodies nivolumab and pembrolizumab. Following approval of ipilimumab in 2011, the latter two achieved market authorization in the summer of 2015. Immune responses thus induced and enhanced inevitably entail autoimmune phenomena, affecting various organs to varying degrees. Knowledge of these side effects is crucial with regard to prevention and management by treating physicians. Typically occurring early on and presenting with pronounced and persistent diarrhea, colitis represents a major and severe side effect. Other immune-mediated disorders include dermatitis, hypophysitis, thyroiditis, hepatitis, iridocyclitis as well as other less common autoimmune phenomena. Early recognition and initiation of treatment can reduce risks and sequelae for patients. This review describes the mechanisms of action of immune checkpoint blockade as well as its clinical effects in metastatic melanoma, with a detailed focus on the spectrum of adverse events and their therapeutic management.
Collapse
Affiliation(s)
- Katharina C Kähler
- Department of Dermatology, Venereology, and Allergology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jessica C Hassel
- Department of Dermatology, and National Cancer Center, University Hospital Heidelberg, Heidelberg, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Carmen Loquai
- Department of Dermatology, Medical Faculty, University of Mainz, Mainz, Germany
| | - Rotraut Mössner
- Department of Dermatology, Venereology, and Allergology, University Medicine Göttingen, Göttingen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Ralf Gutzmer
- Hanover Skin Cancer Center, Department of Dermatology, Venereology, and Allergology, Hanover Medical College, Hanover, Germany
| | | |
Collapse
|
212
|
Vola M, Mónaco A, Bascuas T, Rimsky G, Agorio CI, Chabalgoity JA, Moreno M. TLR7 agonist in combination with Salmonella as an effective antimelanoma immunotherapy. Immunotherapy 2018; 10:665-679. [PMID: 29562809 DOI: 10.2217/imt-2017-0188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM We evaluated a novel approach combining the use of attenuated Salmonella immunotherapy with a Toll-like receptor agonist, imiquimod, in B16F1 melanoma-bearing mice. MATERIALS & METHODS B16F1 melanoma-bearing mice were daily treated with topical imiquimod in combination with one intratumoral injection of attenuated Salmonella enterica serovar Typhimurium LVR01. RESULTS The combined therapy resulted in retarded tumor growth and prolonged survival. Combination treatment led to an enhancement in the expression of pro-inflammatory cytokines and chemokines in the tumor microenvironment, with a Th1-skewed profile, resulting in a broad antitumor response. The induced immunity was effective in controlling the occurrence of metastasis. CONCLUSION Salmonella LVR01 immunotherapy in combination with imiquimod is a novel approach that could be considered as an effective antimelanoma therapy.
Collapse
Affiliation(s)
- Magdalena Vola
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay.,Cátedra de Dermatología, Hospital de Clínicas 'Dr. Manuel Quintela'. Facultad de Medicina, Universidad de la República. Av. Italia s/n, Montevideo, Uruguay
| | - Amy Mónaco
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay
| | - Thais Bascuas
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay
| | - Geraldine Rimsky
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay
| | - Caroline Isabel Agorio
- Cátedra de Dermatología, Hospital de Clínicas 'Dr. Manuel Quintela'. Facultad de Medicina, Universidad de la República. Av. Italia s/n, Montevideo, Uruguay
| | - José Alejandro Chabalgoity
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay
| | - María Moreno
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene. Facultad de Medicina, Universidad de la República. Av. A. Navarro 3051, Montevideo, CP 11600, Uruguay
| |
Collapse
|
213
|
Mirza S, Hill E, Ludlow SP, Nanjappa S. Checkpoint inhibitor-associated drug reaction with eosinophilia and systemic symptom syndrome. Melanoma Res 2018; 27:271-273. [PMID: 28146044 DOI: 10.1097/cmr.0000000000000326] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Drug reaction with eosinophilia and systemic symptom syndrome is a potentially fatal drug reaction that must be recognized quickly. Ipilimumab and nivolumab are both important agents in the treatment of melanoma and continue to be studied in other malignancies. We believe the mainstay of therapy for immunotherapy-induced drug reaction with eosinophilia and systemic symptom syndrome is early recognition, discontinuation of the inciting agent, supportive care, and treatment with high dose corticosteroids with appropriate tapers that may reduce the length of internal organ injury in cases with liver or kidney involvement.
Collapse
Affiliation(s)
- Sayeef Mirza
- aDepartment of Internal Medicine, University of South Florida, Tampa, Florida, USA bMedicine, H. Lee Moffitt Cancer Center and Research Institute Departments of cPharmacy
| | | | | | | |
Collapse
|
214
|
Da Gama Duarte J, Parakh S, Andrews MC, Woods K, Pasam A, Tutuka C, Ostrouska S, Blackburn JM, Behren A, Cebon J. Autoantibodies May Predict Immune-Related Toxicity: Results from a Phase I Study of Intralesional Bacillus Calmette-Guérin followed by Ipilimumab in Patients with Advanced Metastatic Melanoma. Front Immunol 2018; 9:411. [PMID: 29552014 PMCID: PMC5840202 DOI: 10.3389/fimmu.2018.00411] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/14/2018] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced melanoma. The first ICI to demonstrate clinical benefit, ipilimumab, targets cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4); however, the long-term overall survival is just 22%. More than 40 years ago intralesional (IL) bacillus Calmette-Guérin (BCG), a living attenuated strain of Mycobacterium bovis, was found to induce tumor regression by stimulating cell-mediated immunity following a localized and self-limiting infection. We evaluated these two immune stimulants in combination with melanoma with the aim of developing a more effective immunotherapy and to assess toxicity. In this phase I study, patients with histologically confirmed stage III/IV metastatic melanoma received IL BCG injection followed by up to four cycles of intravenous ipilimumab (anti-CTLA-4) (ClinicalTrials.gov number NCT01838200). The trial was discontinued following treatment of the first five patients as the two patients receiving the escalation dose of BCG developed high-grade immune-related adverse events (irAEs) typical of ipilimumab monotherapy. These irAEs were characterized in both patients by profound increases in the repertoire of autoantibodies directed against both self- and cancer antigens. Interestingly, the induced autoantibodies were detected at time points that preceded the development of symptomatic toxicity. There was no overlap in the antigen specificity between patients and no evidence of clinical responses. Efforts to increase response rates through the use of novel immunotherapeutic combinations may be associated with higher rates of irAEs, thus the imperative to identify biomarkers of toxicity remains strong. While the small patient numbers in this trial do not allow for any conclusive evidence of predictive biomarkers, the observed changes warrant further examination of autoantibody repertoires in larger patient cohorts at risk of developing irAEs during their course of treatment. In summary, dose escalation of IL BCG followed by ipilimumab therapy was not well tolerated in advanced melanoma patients and showed no evidence of clinical benefit. Measuring autoantibody responses may provide early means for identifying patients at risk from developing severe irAEs during cancer immunotherapy.
Collapse
Affiliation(s)
- Jessica Da Gama Duarte
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Sagun Parakh
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Miles C Andrews
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia.,MD Anderson Cancer Center, University of Texas, Houston, TX, United States
| | - Katherine Woods
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Anupama Pasam
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Candani Tutuka
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Simone Ostrouska
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Sengenics Corporation, Singapore, Singapore
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| |
Collapse
|
215
|
The role of interim 18F-FDG PET/CT in prediction of response to ipilimumab treatment in metastatic melanoma. Eur J Nucl Med Mol Imaging 2018; 45:1289-1296. [PMID: 29478079 DOI: 10.1007/s00259-018-3972-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE The aim of the present study was to assess the value of interim 18F-FDG PET/CT performed after the first two cycles of ipilimumab treatment in the prediction of the final clinical response to this type of immunotherapy. METHODS The study group comprised 41 patients with unresectable metastatic melanoma scheduled for ipilimumab therapy. Whole-body 18F-FDG PET/CT was performed before the start of ipilimumab treatment (baseline PET/CT) and after the initial two cycles of ipilimumab treatment (interim PET/CT). Evaluation of patient response to treatment was based on the European Organization for Research and Treatment of Cancer (EORTC) 1999 criteria for PET as well as the recently proposed PET Response Evaluation Criteria for Immunotherapy (PERCIMT). The patients' best clinical response, assessed at a median of 21.4 months (range 6.3-41.9 months) was used as reference. RESULTS According to their best clinical response, the patients were divided into two groups: those showing clinical benefit (CB) including stable disease, partial response and complete response (31 patients), and those showing no clinical benefit (no-CB including progressive disease (10 patients). According to the EORTC criteria, interim PET/CT demonstrated progressive metabolic disease (PMD) in 20 patients, stable metabolic disease (SMD) in 11 patients, partial metabolic response (PMR) in 8 patients, and complete metabolic response (CMR) in 2 patients. According to the PERCIMT, interim PET/CT demonstrated PMD in 9 patients, SMD in 24 patients, PMR in 6 patients and CMR in 2 patients. On the basis of the interim PET, the patients were divided in a similar manner to the division according to clinical response into those showing metabolic benefit (MB) including SMD, PMR and CMR, and those showing no metabolic benefit (no-MB) including PMD. According to this dichotomization, the EORTC criteria showed a sensitivity (correctly predicting CB) of 64.5%, a specificity (correctly predicting no-CB) of 90.0%, a positive predictive value (PPV) of 95.2%, a negative predictive value (NPV) of 45.0% and an accuracy of 70.7% in predicting best clinical response. The PERCIMT showed a sensitivity of 93.6%, a specificity of 70.0%, a PPV of 90.6%, a NPV of 77.8% and an accuracy of 87.8%. The McNemar test showed that the PERCIMT had a significantly higher sensitivity than EORTC criteria (p = 0.004), while there was no significant difference in specificity (p = 0.5). The agreement between the two sets of criteria was poor (McNemar test p = 0.001, and accordingly kappa = 0.46). CONCLUSION The application of the recently proposed PERCIMT to interim 18F-FDG PET/CT provides a more sensitive predictor of final clinical response to immunotherapy than the application of the EORTC criteria in patients with metastatic melanoma.
Collapse
|
216
|
Anti-programmed cell death protein 1 tolerance and efficacy after ipilimumab immunotherapy: observational study of 39 patients. Melanoma Res 2018; 27:110-115. [PMID: 27926587 DOI: 10.1097/cmr.0000000000000313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In patients with ipilimumab (IPI)-refractory melanoma, the anti-programmed cell death proteins 1 (PD1s) nivolumab (NIV) and pembrolizumab (PEM) are considered to be a new standard of treatment. Few data are available on anti-PD1 safety in patients who develop IPI-related severe adverse events (AEs) (grade≥3). The aim of this study was to compare the anti-PD1 safety and efficacy in patients with previous severe toxicity to IPI versus in those showing moderate and no previous IPI-related AEs. This single institution-based observational study included all patients treated with anti-PD1 (PEM or NIV) and previously treated with IPI for unresectable stage III or IV melanoma. The patients enrolled were classified according to the occurrence of IPI-related AEs: group A: no previous IPI-related AEs; group B: mild to moderate IPI-related AEs; and group C: severe to life-threatening IPI-related AEs. The main outcome measure was safety of the anti-PD1 among the three groups. The secondary endpoints included response parameters. Groups A, B, and C included, respectively, 16, 13, and 10 patients. The incidence of severe anti-PD1-related AEs (grades 3-4) was 12, 23, and 10% in groups A, B, and C, respectively. One-year estimates of survival were 52.2, 73.4, and 66.7% among the patients in groups A, B, and C, respectively. The number of patients was too small to enable a meaningful statistical comparison. We did not observe any difference in anti-PD1 toxicity onset incidence according to the occurrence of previous IPI AEs. These reassuring real-life data should be confirmed in a wider analysis.
Collapse
|
217
|
Toxicity profile of approved anti-PD-1 monoclonal antibodies in solid tumors: a systematic review and meta-analysis of randomized clinical trials. Oncotarget 2018; 8:8910-8920. [PMID: 27852042 PMCID: PMC5352453 DOI: 10.18632/oncotarget.13315] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/13/2016] [Indexed: 12/13/2022] Open
Abstract
Purpose Nivolumab and pembrolizumab are antibodies against the programmed-death-receptor- 1 (PD-1) which are associated with distinct immune related adverse effects (AEs). This meta-analysis of randomized clinical trials aims to summarize current knowledge regarding the toxicity profile of these agents. Methods PubMed search was conducted in February of 2016. The randomized trials needed to have at least one of the study arms consisting of nivolumab or pembrolizumab monotherapy and a control arm containing no anti-PD-1 therapy. Data were analyzed using random effects meta-analysis for risk ratios. Heterogeneity across studies was analyzed using Q and I2 statistics. Results Nine randomized trials and 5,353 patients were included in our meta-analysis. There was evidence of significant heterogeneity between studies. The pooled relative risk (RR) for treatment-related all grade AEs and grade 3/4 AEs was 0.88 (95% CI 0.81-0.95;P=0.002) and 0.39 (95% CI 0.29-0.53; P<0.001) respectively favoring anti-PD-1 therapy versus standard of care approach. The RR of treatment-related death was 0.45 (95% CI 0.19-1.09; P=0.076). Patients treated with PD-1 inhibitors had an increased risk of hyperthyroidism [RR of 3.44 (95% CI 1.98-5.99; P<0.001)] and hypothyroidism [RR of 6.79 (95% CI 3.10-14.84; P<0.001)]. All grade pruritus and vitiligo were also more common among these patients. The pooled absolute risks of pneumonitis and hypophysitis were 2.65% and 0.47% respectively. Conclusion Approved PD-1 inhibitors are well tolerated, associated with significant low risk of severe treatment-related AEs and increased risk of thyroid dysfunction, pruritus, and vitiligo.
Collapse
|
218
|
A multidisciplinary approach to toxicity management of modern immune checkpoint inhibitors in cancer therapy. Melanoma Res 2018; 26:469-80. [PMID: 27306502 DOI: 10.1097/cmr.0000000000000273] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immune-related Adverse Events (irAEs) are the most significant toxicities associated with the use of checkpoint inhibitors, and result from disinhibition of the host's immune homeostasis. The adverse effects experienced from immunotherapy are significantly different from those of chemotherapy and, to a lesser extent, targeted therapy. Early recognition and diagnosis of these toxicities is often challenging, but is critically important because of the potentially life-threatening nature and associated morbidity. Gastrointestinal, dermatologic, endocrine, and liver toxicities are the most commonly observed. Less commonly, the eyes, pancreas, kidneys, lungs, bone marrow, or nervous system may be affected. Although most irAEs may resolve with supportive care or discontinuation of drug, in severe cases, they may require hospitalization and immune suppressants, such as steroids, and/or may even cause death. The management of immune-related side effects requires a multidisciplinary approach.
Collapse
|
219
|
Lisberg A, Tucker DA, Goldman JW, Wolf B, Carroll J, Hardy A, Morris K, Linares P, Adame C, Spiegel ML, Wells C, McKenzie J, Ledezma B, Mendenhall M, Abarca P, Bornazyan K, Hunt J, Moghadam N, Chong N, Nameth D, Marx C, Madrigal J, Vangala S, Shaverdian N, Elashoff D, Garon EB. Treatment-Related Adverse Events Predict Improved Clinical Outcome in NSCLC Patients on KEYNOTE-001 at a Single Center. Cancer Immunol Res 2018; 6:288-294. [PMID: 29382669 DOI: 10.1158/2326-6066.cir-17-0063] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/19/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022]
Abstract
We retrospectively analyzed non-small cell lung cancer (NSCLC) patients from a single center treated with pembrolizumab on the KEYNOTE-001 trial and evaluated the association between treatment-related adverse events (trAEs) and clinical outcomes. Investigators reported AEs on trial and graded them according to Common Terminology Criteria for Adverse Events v4.0, labeling them as unlikely, possibly, or probably treatment-related. AEs labeled as possibly/probably related were considered trAEs for this analysis. The relationship between the incidence of a trAE and clinical outcomes was evaluated. Ninety-seven NSCLC patients treated on KEYNOTE-001 at the University of California, Los Angeles were evaluated. Ten percent (85/826) of AEs were trAEs, occurring in 40% (39/97) of patients. The most frequent trAEs were rash (21% patients), fatigue (6% patients), and hypothyroidism (6% patients). The 39 patients that experienced a trAE had increased objective response rate (ORR, 38.5%), progression-free survival (PFS: median, 248 days), and overall survival (OS: median, 493 days), compared with the 58 patients that did not (ORR: 8.9%, PFS: median 60 days, OS: median 144.5 days). The observed association between trAEs and improved clinical outcome persisted when using Cox proportional hazards regression models to assess the confounding effect of covariates and mitigate guarantee-time bias. The association also remained when data were substratified by grade, degree of association, and treatment-related select AE designation. This single-center analysis revealed that trAEs predicted for improved clinical outcome with pembrolizumab, and when controlling for guarantee-time bias and plausible confounders, this association remained. This observed relationship adds to our understanding of anti-PD-1 therapy and could aid clinicians in identifying patients most likely to benefit from therapy. Cancer Immunol Res; 6(3); 288-94. ©2018 AACR.
Collapse
Affiliation(s)
- Aaron Lisberg
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - D Andrew Tucker
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Brian Wolf
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - James Carroll
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ariana Hardy
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Karolyn Morris
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Paulina Linares
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Carlos Adame
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Courtney Wells
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jordan McKenzie
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Blanca Ledezma
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Phillip Abarca
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Krikor Bornazyan
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jaime Hunt
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Nima Moghadam
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Natalie Chong
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Danielle Nameth
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Caitlin Marx
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - John Madrigal
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sitaram Vangala
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Narek Shaverdian
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - David Elashoff
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Edward B Garon
- David Geffen School of Medicine at UCLA, Los Angeles, California.
| |
Collapse
|
220
|
Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 2018; 378:158-168. [PMID: 29320654 DOI: 10.1056/nejmra1703481] [Citation(s) in RCA: 3078] [Impact Index Per Article: 439.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Michael A Postow
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| | - Robert Sidlow
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| | - Matthew D Hellmann
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| |
Collapse
|
221
|
Immune-related Adverse Events of Dendritic Cell Vaccination Correlate With Immunologic and Clinical Outcome in Stage III and IV Melanoma Patients. J Immunother 2018; 39:241-8. [PMID: 27227325 PMCID: PMC4902323 DOI: 10.1097/cji.0000000000000127] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The purpose of this study was to determine the toxicity profile of dendritic cell (DC) vaccination in stage III and IV melanoma patients, and to evaluate whether there is a correlation between side effects and immunologic and clinical outcome. This is a retrospective analysis of 82 stage III and 137 stage IV melanoma patients, vaccinated with monocyte-derived or naturally circulating autologous DCs loaded with tumor-associated antigens gp100 and tyrosinase. Median follow-up time was 54.3 months in stage III patients and 12.9 months in stage IV patients. Treatment-related adverse events occurred in 84% of patients; grade 3 toxicity was present in 3% of patients. Most common adverse events were flu-like symptoms (67%) and injection site reactions (50%), and both correlated with the presence of tetramer-positive CD8 T cells (both P<0.001). In stage III melanoma patients experiencing flu-like symptoms, median overall survival (OS) was not reached versus 32.3 months in patients without flu-like symptoms (P=0.009); median OS in patients with an injection site reaction was not reached versus 53.7 months in patients without an injection site reaction (P<0.05). In stage IV melanoma patients (primary uveal and mucosal melanomas excluded), median OS in patients with or without flu-like symptoms was 13.1 versus 8.9 months, respectively (P=0.03); median OS in patients with an injection site reaction was 15.7 months versus 9.8 months in patients without an injection site reaction (P=0.003). In conclusion, DC vaccination is safe and tolerable and the occurrence of the immune-related side effects, such as flu-like symptoms and injection site reactions, correlates with immunologic and clinical outcome.
Collapse
|
222
|
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018; 131:58-67. [PMID: 29118008 PMCID: PMC6317697 DOI: 10.1182/blood-2017-06-741033] [Citation(s) in RCA: 816] [Impact Index Per Article: 116.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 02/08/2023] Open
Abstract
CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
223
|
Melanoma: Immunotherapy in Advanced Melanoma and in the Adjuvant Setting. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
224
|
Patel AB, Pacha O. Skin Reactions to Immune Checkpoint Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 995:117-129. [DOI: 10.1007/978-3-030-02505-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
225
|
Abstract
Cancer immunotherapies, widely heralded as transformational for many adult cancer patients, are becoming viable options for selected subsets of pediatric cancer patients. Many therapies are currently being investigated, from immunomodulatory agents to adoptive cell therapy, bispecific T-cell engagers, oncolytic virotherapy, and checkpoint inhibition. One of the most exciting immunotherapies recently FDA approved is the use of CD19 chimeric antigen receptor T cells for pre-B-cell acute lymphoblastic leukemia. With this approval and others, immunotherapy for pediatric cancers is gaining traction. One of the caveats to many of these immunotherapies is the challenge of predictive biomarkers; determining which patients will respond to a given therapy is not yet possible. Much research is being focused on which biomarkers will be predictive and prognostic for these patients. Despite many benefits of immunotherapy, including less long-term side effects, some treatments are fraught with immediate side effects that range from mild to severe, although most are manageable. With few downsides and the potential for disease cures, immunotherapy in the pediatric population has the potential to move to the front-line of therapeutic options.
Collapse
Affiliation(s)
- Mary Frances Wedekind
- 0000 0001 2285 7943grid.261331.4Division of Pediatric Hematology/Oncology/Bone and Marrow Transplant, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205 USA ,0000 0001 2285 7943grid.261331.4Center for Childhood Cancer and Blood Disorders, The Research Institute, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Research Bldg II, Columbus, OH 43205 USA
| | - Nicholas L. Denton
- 0000 0001 2285 7943grid.261331.4Center for Childhood Cancer and Blood Disorders, The Research Institute, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Research Bldg II, Columbus, OH 43205 USA
| | - Chun-Yu Chen
- 0000 0001 2285 7943grid.261331.4Center for Childhood Cancer and Blood Disorders, The Research Institute, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Research Bldg II, Columbus, OH 43205 USA
| | - Timothy P. Cripe
- 0000 0001 2285 7943grid.261331.4Division of Pediatric Hematology/Oncology/Bone and Marrow Transplant, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205 USA ,0000 0001 2285 7943grid.261331.4Center for Childhood Cancer and Blood Disorders, The Research Institute, Nationwide Children’s Hospital, The Ohio State University, 700 Children’s Drive, Research Bldg II, Columbus, OH 43205 USA
| |
Collapse
|
226
|
Iglesias P. Cancer immunotherapy-induced endocrinopathies: Clinical behavior and therapeutic approach. Eur J Intern Med 2018; 47:6-13. [PMID: 28826822 DOI: 10.1016/j.ejim.2017.08.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022]
Abstract
Cancer immunotherapy has proven to be effective in a wide variety of tumors. The use of immune checkpoint blocking monoclonal antibodies has become a standard treatment regimen in some of them as advanced melanoma. However, given the mechanism of action, its use may be associated with immune-related adverse events that may complicate the clinical course and prognosis of patients. Among these are autoimmune endocrine adverse effects, such as hypophysitis, hypo and hyperthyroidism, and adrenal insufficiency. This review focuses on the most relevant and new aspects related to the incidence, clinical presentation, diagnosis and treatment of these adverse effects associated with different types of immune checkpoint inhibitors in cancer immunotherapy.
Collapse
Affiliation(s)
- Pedro Iglesias
- Department of Endocrinology, Hospital Ramón y Cajal, Ctra. de Colmenar, Km 9.100, 28034 Madrid, Spain.
| |
Collapse
|
227
|
McCarthy A, Sheahan K. Pathologic Response of the Gastrointestinal Tract to Toxicants. COMPREHENSIVE TOXICOLOGY 2018:113-138. [DOI: 10.1016/b978-0-12-801238-3.95672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
228
|
Jain A, Shannon VR, Sheshadri A. Immune-Related Adverse Events: Pneumonitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 995:131-149. [PMID: 30539509 DOI: 10.1007/978-3-030-02505-2_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Checkpoint inhibitors are part of the family of immunotherapies and are increasingly being used in a wide variety of cancers. Immune-related adverse events pose a major challenge in the treatment of cancer patients. Pneumonitis is a rare immune-related adverse event that presents in distinct patterns. The goal of this chapter is to instruct readers on the incidence and clinical manifestations of pneumonitis and to offer guidance in the evaluation and treatment of patients with pneumonitis.
Collapse
Affiliation(s)
- Akash Jain
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vickie R Shannon
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
229
|
Festino L, Ascierto PA. Side Effects of Cancer Immunotherapy with Checkpoint Inhibitors. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
230
|
Sakamuri D, Glitza IC, Betancourt Cuellar SL, Subbiah V, Fu S, Tsimberidou AM, Wheler JJ, Hong DS, Naing A, Falchook GS, Fanale MA, Cabanillas ME, Janku F. Phase I Dose-Escalation Study of Anti-CTLA-4 Antibody Ipilimumab and Lenalidomide in Patients with Advanced Cancers. Mol Cancer Ther 2017; 17:671-676. [PMID: 29237802 DOI: 10.1158/1535-7163.mct-17-0673] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/10/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022]
Abstract
Preclinical data suggest that combining a checkpoint inhibition with immunomodulatory derivative can increase anticancer response. We designed a dose-escalation study using a 3 + 3 design to determine the safety, maximum tolerated dose (MTD) or recommended phase II dose (R2PD) and dose-limiting toxicities (DLT) of the anti-CTLA-4 antibody ipilimumab (1.5-3 mg/kg intravenously every 28 days × 4) and lenalidomide (10-25 mg orally daily for 21 of 28 days until disease progression or unacceptable toxicity) in advanced cancers. Total of 36 patients (Hodgkin lymphoma, 7; melanoma, 5; leiomyosarcoma, 4; renal cancer, 3; thyroid cancer, 3; other cancers, 14; median of 3 prior therapies) were enrolled. The MTD has not been reached and ipilimumab 3 mg/kg and lenalidomide 25 mg have been declared as R2PD. DLT were grade (G) 3 rash (3 patients) and G3 pancreatitis (1 patient). G3/4 drug-related toxicities other than DLT were G3 anemia (5 patients), G3 thromboembolism (2 patients), G3 thrombocytopenia, G3 rash, G3 hypopituitarism, G3 pneumonitis, G3 transaminitis, and G4 hypopituitarism (all in 1 patient). Eight patients had tumor shrinkage per immune-related response criteria (-79% to -2%) including a PR (-79% for 7.2+ months) in a refractory Hodgkin lymphoma. Using comprehensive genomic profiling, a total mutation burden (mutations/Mb) was evaluated in 17 patients, with one of the patients achieving a PR demonstrated intermediate mutation burden. In conclusion, combination of ipilimumab and lenalidomide is well tolerated and demonstrated preliminary signals of activity in patients with refractory Hodgkin lymphoma and other advanced cancers. Mol Cancer Ther; 17(3); 671-6. ©2017 AACR.
Collapse
Affiliation(s)
- Divya Sakamuri
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer J Wheler
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gerald S Falchook
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.,Sarah Cannon Research Institute at HealthONE, Denver, Colorado
| | - Michelle A Fanale
- Department of Lymphoma and Myeloma Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
231
|
Mo X, Zhang H, Preston S, Martin K, Zhou B, Vadalia N, Gamero AM, Soboloff J, Tempera I, Zaidi MR. Interferon-γ Signaling in Melanocytes and Melanoma Cells Regulates Expression of CTLA-4. Cancer Res 2017; 78:436-450. [PMID: 29150430 DOI: 10.1158/0008-5472.can-17-1615] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/19/2017] [Accepted: 11/01/2017] [Indexed: 12/18/2022]
Abstract
CTLA4 is a cell surface receptor on T cells that functions as an immune checkpoint molecule to enforce tolerance to cognate antigens. Anti-CTLA4 immunotherapy is highly effective at reactivating T-cell responses against melanoma, which is postulated to be due to targeting CTLA4 on T cells. Here, we report that CTLA4 is also highly expressed by most human melanoma cell lines, as well as in normal human melanocytes. Interferon-γ (IFNG) signaling activated the expression of the human CTLA4 gene in a melanocyte and melanoma cell-specific manner. Mechanistically, IFNG activated CTLA4 expression through JAK1/2-dependent phosphorylation of STAT1, which bound a specific gamma-activated sequence site on the CTLA4 promoter, thereby licensing CBP/p300-mediated histone acetylation and local chromatin opening. In melanoma cell lines, elevated baseline expression relied upon constitutive activation of the MAPK pathway. Notably, RNA-seq analyses of melanoma specimens obtained from patients who had received anti-CTLA4 immunotherapy (ipilimumab) showed upregulation of an IFNG-response gene expression signature, including CTLA4 itself, which correlated significantly with durable response. Taken together, our results raise the possibility that CTLA4 targeting on melanoma cells may contribute to the clinical immunobiology of anti-CTLA4 responses.Significance: These findings show that human melanoma cells express high levels of the immune checkpoint molecule CTLA4, with important possible implications for understanding how anti-CTLA4 immunotherapy mediates its therapeutic effects. Cancer Res; 78(2); 436-50. ©2017 AACR.
Collapse
Affiliation(s)
- Xuan Mo
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Hanghang Zhang
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Sarah Preston
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kayla Martin
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Bo Zhou
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Nish Vadalia
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ana M Gamero
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania.,Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Italo Tempera
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania.,Department of Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - M Raza Zaidi
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania. .,Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
232
|
Konda B, Nabhan F, Shah MH. Endocrine dysfunction following immune checkpoint inhibitor therapy. Curr Opin Endocrinol Diabetes Obes 2017; 24:337-347. [PMID: 28661915 DOI: 10.1097/med.0000000000000357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICI) represent an important milestone in the modern era of antineoplastic therapy and have ushered optimism amongst oncologists and patients alike. These agents, however, are associated with significant potential toxicities, the importance of which cannot be overstated. The clinical presentation, diagnosis, and management strategies of immune-related endocrinopathies associated with ICI use are described in this case-based review. RECENT FINDINGS An increasing number of ICI have shown promise in the management of various malignancies in the recent years. These include cytotoxic T lymphocyte antigen-4 inhibitors, programmed cell death 1 (PD-1) antibodies, and PD-ligand 1 (PD-L1) antibodies. Several endocrinopathies, including hypophysitis, thyroid dysfunction, hyperglycemia, and primary adrenal insufficiency, have been associated with the use of these agents. Toxicities may range from mild transient laboratory abnormalities to potentially life-threatening ones, warranting immediate therapeutic intervention. Combination ICI therapies may be associated with a greater risk of endocrine dysfunction when compared with monotherapy. The clinical presentation and laboratory assessment of these patients often pose a diagnostic challenge as they may be confused by the symptoms related to their underlying malignancy or potential associated acute illnesses. SUMMARY ICI use is associated with serious endocrinopathies that may have a nonspecific initial presentation. A constant eye for these symptoms and a systematic approach to diagnosis are essential for prompt initiation of therapy and prevention of significant complications.
Collapse
Affiliation(s)
- Bhavana Konda
- aDivision of Medical Oncology, Department of Internal Medicine bDivision of Endocrinology, Diabetes, and Metabolism, The Ohio State University and Arthur G. James Cancer Center, Columbus, Ohio, USA
| | | | | |
Collapse
|
233
|
de Coaña YP, Wolodarski M, Poschke I, Yoshimoto Y, Yang Y, Nyström M, Edbäck U, Brage SE, Lundqvist A, Masucci GV, Hansson J, Kiessling R. Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long-term survivors with advanced melanoma. Oncotarget 2017; 8:21539-21553. [PMID: 28423487 PMCID: PMC5400604 DOI: 10.18632/oncotarget.15368] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022] Open
Abstract
Ipilimumab has revolutionized malignant melanoma therapy, but a better understanding of the mechanisms behind treatment response and adverse effects is needed. In this work, the immune system of ipilimumab treated patients was monitored to investigate potential mechanisms of action that may correlate with treatment outcome. Blood samples from 43 advanced melanoma patients were taken before, during and at the end of treatment. Hematological parameters were measured and flow cytometry analysis was performed in fresh samples within two hours of sample collection. Strong differences in markers CD45RA, CCR7, HLA-DR and CD15 between fresh and cryopreserved samples were observed. Ipilimumab treatment increased absolute lymphocyte counts, eosinophils, effector T cells and their activation status, whilst diminishing the suppressive side of the immune response, acting on regulatory T cells and myeloid derived suppressor cells (MDSCs). These effects were visible after one ipilimumab infusion and, regarding eosinophil counts, correlated with onset of adverse events. Monocytic MDSCs were decreased in response to treatment only in patients with clinical benefit; additionally, patients with a lower frequency of these cells after the first ipilimumab infusion experienced increased overall survival. CD8 effector memory T cell frequencies at the end of treatment were higher in patients with clinical benefit and positively correlated with survival. These data show that a clinical response to ipilimumab not only requires reshaping T cell populations, but additionally involves a reduction in suppressive cells such as monocytic MDSCs. Our work could provide insight on predicting treatment outcome, assisting clinicians in offering the best personalized therapeutic approach.
Collapse
Affiliation(s)
- Yago Pico de Coaña
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Maria Wolodarski
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital Solna, Stockholm, Sweden
| | - Isabel Poschke
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Yuya Yoshimoto
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuan Yang
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Cancer Immunology and Immunotherapy Center, The Affiliated Hospital of Guiyang Medical College, Guiyang, People's Republic of China
| | - Maria Nyström
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Edbäck
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Suzanne Eghyazi Brage
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Cell Therapies Institute, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Giuseppe V Masucci
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital Solna, Stockholm, Sweden
| | - Johan Hansson
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital Solna, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
234
|
Torres-García D, Pérez-Torres A, Manoutcharian K, Orbe U, Servín-Blanco R, Fragoso G, Sciutto E. GK-1 peptide reduces tumor growth, decreases metastatic burden, and increases survival in a murine breast cancer model. Vaccine 2017; 35:5653-5661. [PMID: 28890195 DOI: 10.1016/j.vaccine.2017.08.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/10/2017] [Accepted: 08/19/2017] [Indexed: 12/31/2022]
Abstract
GK-1 is a parasite-derived peptide adjuvant of 18 amino acid-length that enhances T-cell function and increases survival in B16-F10 melanoma tumor-bearing mice. This study was designed to evaluate in vivo the antitumor efficacy of GK-1 on 4T1 mouse mammary carcinoma. BALB/c mice with palpable primary tumors were weekly intravenously injected three times with saline solution or three different concentrations (10, 50, or 100μg per mouse) of GK-1. GK-1 significantly increased lifespan (p<0.0001) and reduced the primary tumor weight (p=0.014) and volume (p<0.0001) with respect to control mice, with no statistically significant differences among GK-1 doses. At the primary tumor, we found increased necrotic areas associated with a reduction in tumor mass, as well as an increase in the antitumor cytokine IL-12. Especially encouraging is the ability of GK-1 to reduce the number of lung metastasis (p=0.006) disregarding the dose used. The participation of IL-6 in metastasis development and the decreased levels of CCL-2, CCL-3, TNF-α, CXCL-9, GM-CSF, and b-FGF found in lungs of GK-1-treated mice is discussed. Our study supports the effectiveness of GK-1 as an antineoplastic agent that merits further exploration in combination with other therapeutic approaches in future translational studies.
Collapse
Affiliation(s)
- D Torres-García
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - A Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - K Manoutcharian
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - U Orbe
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - R Servín-Blanco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - G Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - E Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico.
| |
Collapse
|
235
|
Pérez-De-Lis M, Retamozo S, Flores-Chávez A, Kostov B, Perez-Alvarez R, Brito-Zerón P, Ramos-Casals M. Autoimmune diseases induced by biological agents. A review of 12,731 cases (BIOGEAS Registry). Expert Opin Drug Saf 2017; 16:1255-1271. [DOI: 10.1080/14740338.2017.1372421] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Marta Pérez-De-Lis
- Servicio de Anestesiologia y Reanimación, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Soledad Retamozo
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Hospital Privado Universitario de Córdoba, Córdoba, Argentina
- Instituto De Investigaciones En Ciencias De La Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Alejandra Flores-Chávez
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Unidad de Investigación Biomédica 02, Unidad de Investigación en Epidemiología Clínica, Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Hospital de Especialidades, Guadalajara, Mexico
- Programa de Doctorado en Ciencias Médicas, Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, Mexico
| | | | | | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA- Sanitas, Barcelona, Spain
| | - Manuel Ramos-Casals
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
236
|
Rawson RV, Robbins E, Kapoor R, Scolyer RA, Long GV. Recurrent bowel obstruction: unusual presentation of pembrolizumab-induced pancreatitis in annular pancreas. Eur J Cancer 2017; 82:167-170. [DOI: 10.1016/j.ejca.2017.05.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 12/31/2022]
|
237
|
Kidwell KM, Postow MA, Panageas KS. Sequential, Multiple Assignment, Randomized Trial Designs in Immuno-oncology Research. Clin Cancer Res 2017; 24:730-736. [PMID: 28835379 DOI: 10.1158/1078-0432.ccr-17-1355] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/03/2017] [Accepted: 08/17/2017] [Indexed: 01/13/2023]
Abstract
Clinical trials investigating immune checkpoint inhibitors have led to the approval of anti-CTLA-4 (cytotoxic T-lymphocyte antigen-4), anti-PD-1 (programmed death-1), and anti-PD-L1 (PD-ligand 1) drugs by the FDA for numerous tumor types. In the treatment of metastatic melanoma, combinations of checkpoint inhibitors are more effective than single-agent inhibitors, but combination immunotherapy is associated with increased frequency and severity of toxicity. There are questions about the use of combination immunotherapy or single-agent anti-PD-1 as initial therapy and the number of doses of either approach required to sustain a response. In this article, we describe a novel use of sequential, multiple assignment, randomized trial (SMART) design to evaluate immune checkpoint inhibitors to find treatment regimens that adapt within an individual based on intermediate response and lead to the longest overall survival. We provide a hypothetical example SMART design for BRAF wild-type metastatic melanoma as a framework for investigating immunotherapy treatment regimens. We compare implementing a SMART design to implementing multiple traditional randomized clinical trials. We illustrate the benefits of a SMART over traditional trial designs and acknowledge the complexity of a SMART. SMART designs may be an optimal way to find treatment strategies that yield durable response, longer survival, and lower toxicity. Clin Cancer Res; 24(4); 730-6. ©2017 AACR.
Collapse
Affiliation(s)
- Kelley M Kidwell
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, Michigan.
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Katherine S Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
238
|
Rodríguez JA. HLA-mediated tumor escape mechanisms that may impair immunotherapy clinical outcomes via T-cell activation. Oncol Lett 2017; 14:4415-4427. [PMID: 29085437 PMCID: PMC5649701 DOI: 10.3892/ol.2017.6784] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 03/31/2017] [Indexed: 12/15/2022] Open
Abstract
Although the immune system provides protection from cancer by means of immunosurveillance, which serves a major function in eliminating cancer cells, it may also lead to cancer immunoediting, molding tumor immunogenicity. Cancer cells exploit several molecular mechanisms to thwart immune-mediated death by disabling cellular components of the immune system associated with tumor recognition and rejection. Human leukocyte antigen (HLA) molecules are mandatory for the immune recognition and subsequent killing of neoplastic cells by the immune system, as tumor antigens must be presented in an HLA-restricted manner to be recognized by T-cell receptors. Impaired HLA-I expression prevents the activation of cytotoxic immune mechanisms, whereas impaired HLA-II expression affects the antigen-presenting capability of antigen presenting cells. Aberrant HLA-G expression by cancer cells favors immune escape by inhibiting the activities of virtually all immune cells. The development of cancer therapies based on T-cell activation must consider these HLA-associated immune evasion mechanisms, as alterations in their expression occur early and frequently in the majority of types of cancer, and have an adverse impact on the clinical response to immunotherapy. Herein, the concept of altered HLA expression as a mechanism exploited by tumors to escape immune control and induce an immunosuppressive environment is reviewed. A number of novel clinical immunotherapeutic approaches used for cancer treatment are also reviewed, and strategies for overcoming the limitations of these immunotherapeutic interventions are proposed.
Collapse
Affiliation(s)
- Josefa A Rodríguez
- Cancer Biology Research Group, National Cancer Institute of Colombia, 111511 Bogotá, Colombia
| |
Collapse
|
239
|
Chang ST, Menias CO, Lubner MG, Mellnick VM, Hara AK, Desser TS. Molecular and Clinical Approach to Intra-abdominal Adverse Effects of Targeted Cancer Therapies. Radiographics 2017; 37:1461-1482. [PMID: 28753381 DOI: 10.1148/rg.2017160162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeted cancer therapies encompass an exponentially growing number of agents that involve a myriad of molecular pathways. To excel within this rapidly changing field of clinical oncology, radiologists must eschew traditional organ system-based approaches of cataloging adverse effects in favor of a conceptual framework that incorporates molecular mechanisms and associated clinical outcomes. Understanding molecular mechanisms that underlie imaging manifestations of adverse effects and known associations with treatment response allows radiologists to more effectively recognize adverse effects and differentiate them from tumor progression. Radiologists can therefore more effectively guide oncologists in the management of adverse effects and treatment decisions regarding continuation or cessation of drug therapy. Adverse effects from targeted cancer therapies can be classified into four categories: (a) category 1, on-target adverse effects associated with treatment response; (b) category 2, on-target adverse effects without associated treatment response; (c) category 3, off-target adverse effects; and (d) category 4, tumor necrosis-related adverse effects. This review focuses on adverse effects primarily within the abdomen and pelvis classified according to established or hypothesized molecular mechanisms and illustrated with images of classic examples and several potential emerging toxic effects. ©RSNA, 2017.
Collapse
Affiliation(s)
- Stephanie T Chang
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Christine O Menias
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Meghan G Lubner
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Vincent M Mellnick
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Amy K Hara
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Terry S Desser
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| |
Collapse
|
240
|
Charles J, Chaperot L, Revol B, Baudin M, Mouret S, Hamon A, Leccia MT, Plumas J, Aspord C. The avidity of tumor-specific T cells amplified by a plasmacytoid dendritic cell-based assay can predict the clinical evolution of melanoma patients. Pigment Cell Melanoma Res 2017; 31:82-94. [PMID: 28741900 DOI: 10.1111/pcmr.12618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 07/01/2017] [Indexed: 12/22/2022]
Abstract
The advent of immune checkpoint blockers and targeted therapies has changed the outcome of melanoma. However, many patients experience relapses, emphasizing the need for predictive and prognostic biomarkers. We developed a strategy based on plasmacytoid dendritic cells (pDCs) loaded with melanoma tumor antigens that allows eliciting highly efficient antitumor T-cell responses. We used it to investigate antitumor T-cell functionality in peripheral blood mononuclear cells and tumor-infiltrating lymphocytes from melanoma patients. The pDCs elicited tumor-specific T cells in different proportions and displaying diverse functional features, dependent upon the stage of the disease, but independent of the histological parameters at diagnosis. Strikingly, the avidity of the MelA-specific T cells triggered by the pDCs was found to predict patient relapse time and overall survival. Our findings highlighted unexplored aspects of antitumor T-cell responsiveness in melanoma, and revealed for the first time the structural avidity of tumor-specific T cells as a crucial feature for predicting clinical evolution.
Collapse
Affiliation(s)
- Julie Charles
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,Dermatology, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Laurence Chaperot
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,R&D Laboratory, Etablissement Français du Sang Rhone-Alpes, La Tronche, France
| | - Bruno Revol
- Pharmacovigilance Department, CHU Grenoble Alpes, Grenoble, France
| | - Marine Baudin
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,R&D Laboratory, Etablissement Français du Sang Rhone-Alpes, La Tronche, France
| | - Stephane Mouret
- Dermatology, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Agnes Hamon
- Laboratoire Jean Kuntzmann, Universite Grenoble Alpes, Grenoble, France
| | - Marie-Therese Leccia
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,Dermatology, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Joel Plumas
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,R&D Laboratory, Etablissement Français du Sang Rhone-Alpes, La Tronche, France
| | - Caroline Aspord
- University Grenoble Alpes, Grenoble, France.,Immunobiology& Immunotherapy of Chronic Diseases, U1209, INSERM, La Tronche, France.,R&D Laboratory, Etablissement Français du Sang Rhone-Alpes, La Tronche, France
| |
Collapse
|
241
|
Abstract
PURPOSE OF REVIEW Checkpoint inhibitors have been increasingly considered as new targets for cancer therapies. Patients receiving checkpoint inhibitors develop many immune-related adverse events (IRAEs). However, ophthalmic IRAEs are rare and have been reported in less than 1% of patients. To date, few case reports evaluating the ophthalmological side-effects of checkpoint inhibitors have been published. In this review, we plan to report the different ocular and orbital side-effects of the checkpoint inhibitors, and to help guide ophthalmologists and oncologists in their management. RECENT FINDINGS Ocular side-effects of checkpoint inhibitors include peripheral ulcerative keratitis, uveitis, Vogt-Koyanagi-Harada syndrome, choroidal neovascularization and melanoma-associated retinopathy. Both thyroid-associated orbitopathy and idiopathic orbital inflammation have also been reported in association with checkpoint inhibitors. Mild IRAE can be treated with topical steroids, whereas systemic corticosteroids and discontinuation of checkpoint inhibitors are indicated in more severe ocular and orbital inflammation. SUMMARY Physicians involved in the care of oncologic patients should be aware of the ocular and orbital IRAEs that may develop with checkpoint inhibitors. A strong cooperation between oncologists and ophthalmologists is required in the diagnosis and prompt management of these IRAEs.
Collapse
|
242
|
Abstract
PURPOSE OF REVIEW Three mAbs targeting immune checkpoint proteins are available for the treatment of patients with melanoma, lung, and kidney cancer, and their use will likely expand in the future to additional tumor types. We here update the literature on the incidence and pathophysiology of endocrine toxicities induced by these agents, and discuss management guidance. RECENT FINDINGS Immune checkpoint inhibition may trigger autoimmune syndromes involving different organs, including several endocrine glands (pituitary, thyroid, adrenals, and endocrine pancreas). Hypophysitis is more frequently associated with ipilimumab, whereas the incidence of thyroid dysfunction is higher with nivolumab/pembrolizumab. Primary adrenal insufficiency can rarely occur with either treatment. Autoimmune diabetes is very rare. As hypophysitis and adrenalitis may be life-threatening, endocrinological evaluation is essential particularly in patients developing fatigue and other symptoms consistent with adrenal insufficiency. Corticosteroids should be promptly used when hypophysitis-induced adrenal insufficiency or adrenalitis are diagnosed, but not in thyroiditis or diabetes. No impact of corticosteroids on the efficacy/activity of immune checkpoint-inhibiting drugs is reported. Hormonal deficiencies are often permanent. SUMMARY In absence of predicting factors, accurate information to patients provided by the oncology care team is essential for early diagnosis and to limit the consequences of checkpoint inhibition-related endocrine toxicity.
Collapse
|
243
|
Dick J, Lang N, Slynko A, Kopp-Schneider A, Schulz C, Dimitrakopoulou-Strauss A, Enk AH, Hassel JC. Use of LDH and autoimmune side effects to predict response to ipilimumab treatment. Immunotherapy 2017; 8:1033-44. [PMID: 27485076 DOI: 10.2217/imt-2016-0083] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Ipilimumab is a cytotoxic T-lymphocyte antigen-4 antibody that enhances T-cell activity and proliferation. METHODS In a retrospective analysis of 86 patients the clinical benefits of ipilimumab treatment were correlated with laboratory and clinical data. RESULTS A lactate dehydrogenase (LDH) value within the normal range before the start of therapy was significantly correlated with better OS (p ≤ 0.009). An increase in LDH level after two cycles was indicative of a poor outcome, and was significantly negatively correlated with treatment response and overall survival and progression-free survival. 42% of all patients suffered from autoimmune toxicity (CTCAE grades 2-4). The occurrence of autoimmune toxicity clearly correlated with clinical benefit. CONCLUSION Changes in LDH level and side effects correlate with response to therapy and survival.
Collapse
Affiliation(s)
- J Dick
- Department of Dermatology & National Centre for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - N Lang
- Department of Dermatology & National Centre for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - A Slynko
- German Cancer Research Centre DKFZ, Department of Biostatistics, Heidelberg, Germany
| | - A Kopp-Schneider
- German Cancer Research Centre DKFZ, Department of Biostatistics, Heidelberg, Germany
| | - C Schulz
- Department of Dermatology & National Centre for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | - A H Enk
- Department of Dermatology & National Centre for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - J C Hassel
- Department of Dermatology & National Centre for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
244
|
Moreira A, Leisgang W, Schuler G, Heinzerling L. Eosinophilic count as a biomarker for prognosis of melanoma patients and its importance in the response to immunotherapy. Immunotherapy 2017; 9:115-121. [PMID: 28128709 DOI: 10.2217/imt-2016-0138] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM The prognostic role of eosinophils in cancer has been controversial. Some entities such as gastrointestinal cancers show a better survival, while others such as Hodgkin's lymphoma a worse survival in patients with eosinophilia. Patients who exhibited an increase in eosinophils upon therapy with ipilimumab or pembrolizumab were shown to survive longer. We wanted to investigate whether eosinophilia is a prognostic marker in metastatic melanoma. METHODS In total, 173 patients with metastatic melanoma from our data base (median age 60 years; n = 86 with immunotherapy, n = 87 without immunotherapy) were analyzed for eosinophil counts and survival over the course of 12 years. Eosinophilic count was detected by peripheral blood smear. The ethical committee had approved this retrospective study. RESULTS Melanoma patients with eosinophilia at any point in their course of disease show a trend toward longer survival independently of their therapy. There is a statistically significant difference for the patients who survive at least 12 months (p < 0.005). In patients with checkpoint inhibitor therapy, survival was significantly prolonged in every patient with eosinophilia (p < 0.05). Furthermore, 69% of the patients treated with immunotherapy experienced at least once an eosinophilia of 5% or greater compared with 46% in the immunotherapy naive-group; for an eosinophilia of 10% values were 30 and 9%, respectively. Interestingly, in patients with more than 20% eosinophils (n = 7) survival was prolonged with a median of 35 months (range 19-60 months) as compared with 16 months (range 1-117 months). CONCLUSION Eosinophilia is a prognostic marker in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Waltraud Leisgang
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
245
|
Simonelli M, Di Tommaso L, Baretti M, Santoro A. Pathological characterization of nivolumab-related liver injury in a patient with glioblastoma. Immunotherapy 2017; 8:1363-1369. [PMID: 28000537 DOI: 10.2217/imt-2016-0057] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint inhibitors such as anti-CTLA-4 and anti-PD-1/PD-L1 monoclonal antibodies have dramatically changed the paradigm of cancer therapy over the past few years. The use of these agents is associated with a unique pattern of autoimmune-like/inflammatory side effects termed immune-related adverse events (irAEs), that may cause collateral damage to normal tissues. Although severe irAEs remain rare, they can become life-threatening if not anticipated and managed appropriately. Improving our knowledge of the mechanisms underlying the development of these toxicities is crucial to optimize clinical efficacy and safety of these new immunotherapeutics. Herein we describe for the first time the pathological features of a severe liver-injury associated with the administration of the anti-PD-1 agent nivolumab in a patient with glioblastoma.
Collapse
Affiliation(s)
- Matteo Simonelli
- Humanitas Cancer Center, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Luca Di Tommaso
- Department of Pathology, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Mi), Italy.,Humanitas University, Via Alessandro Manzoni 113, 20089 Rozzano (Mi), Italy
| | - Marina Baretti
- Humanitas Cancer Center, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Armando Santoro
- Humanitas Cancer Center, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Mi), Italy.,Humanitas University, Via Alessandro Manzoni 113, 20089 Rozzano (Mi), Italy
| |
Collapse
|
246
|
Functionality of Patients 75 Years and Older Undergoing Mohs Micrographic Surgery: A Multicenter Study. Dermatol Surg 2017; 43:904-910. [DOI: 10.1097/dss.0000000000001111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
247
|
Limitations and opportunities for immune checkpoint inhibitors in pediatric malignancies. Cancer Treat Rev 2017; 58:22-33. [PMID: 28622628 PMCID: PMC5524462 DOI: 10.1016/j.ctrv.2017.05.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICI) have shown great promise in a wide spectrum of adult solid and hematological malignancies, achieving objective tumor responses and prolonging survival. However, there is limited clinical success amongst pediatric patients. In this review, we summarize the current understanding of ICI and present an up-to-date overview of recent and ongoing clinical trials of ICI in pediatric malignancies. In addition, we will discuss immunologic and clinical difficulties in this young population, as well as future prospects for combination of ICI with other immune-based and conventional treatments.
Collapse
|
248
|
Hasegawa Y, Kawai S, Ota T, Tsukuda H, Fukuoka M. Myasthenia gravis induced by nivolumab in patients with non-small-cell lung cancer: a case report and literature review. Immunotherapy 2017. [PMID: 28649876 DOI: 10.2217/imt-2017-0043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A 76-year-old woman who was diagnosed with non-small-cell lung cancer presented with left eyelid ptosis and grade 4 creatine phosphokinase elevation after the second cycle of nivolumab monotherapy. Nivolumab has demonstrated promising efficacy in patients with non-small-cell lung cancer in several trials. Dyspnea and muscle weakness developed rapidly with an acute exacerbation. She underwent plasmapheresis and intravenous immune globulin followed by treatment with low-dose prednisolone. She had gradual symptoms improvement. We diagnosed her with myasthenia gravis (MG) based on her symptoms and the detection of anti-acetylcholine receptor antibody. According to postmarketing surveillance in 15,740 Japanese patients, the total incidence rate of MG is 0.1%. We report a rare case of drug-induced MG in a patient receiving nivolumab.
Collapse
Affiliation(s)
| | - Shigeru Kawai
- Department of Neurology, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Takayo Ota
- Department of Medical Oncology, Izumi Municipal Hospital, Izumi, Japan
| | - Hiroshi Tsukuda
- Department of Medical Oncology, Izumi Municipal Hospital, Izumi, Japan
| | - Masahiro Fukuoka
- Department of Medical Oncology, Izumi Municipal Hospital, Izumi, Japan
| |
Collapse
|
249
|
Wang W, Lie P, Guo M, He J. Risk of hepatotoxicity in cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis of published data. Int J Cancer 2017; 141:1018-1028. [PMID: 28263392 DOI: 10.1002/ijc.30678] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/14/2017] [Indexed: 12/13/2022]
Abstract
Although existing evidence from clinical trials has demonstrated manifestation of hepatic adverse events with the use of immune checkpoint inhibitors (ICPIs), overall risks have yet to be reported. Therefore, we assessed the risk of hepatotoxicity associated with inhibitors of the immune checkpoint. We examined data from the Pubmed, Medline and Google Scholar databases. We also examined original studies and review articles for crossreferences Eligible studies included randomized Phase II to Phase III trials of cancer patients treated with nivolumab, pembrolizumab, ipilimumab, tremelimumab. The authors extracted relevant information on participants' characteristics, all-grade and high-grade hepatotoxicity and information on the methodology of the studies. In total, 17 trials were considered eligible for the meta-analysis. The odds ratio for all-grade hepatotoxicity for CTLA-4 inhibitors (Ipilimumab and tremelimumab) was 1.24 (95% confidence interval 0.75, 2.05; p = 0.39) and for high-grade hepatotoxicity was 1.93 (95% confidence interval 0.84, 4.44; p =0.12). Moreover, the odds ratio for all-grade hepatotoxicity for PD-1 inhibitors was 1.52 (95% confidence interval 1.24, 1.86; p < 0.0001) and for high-grade hepatotoxicity was 0.48 (95% confidence interval 0.29, 0.80; p =0.005). The analysis of data showed that CTLA-4 inhibitors seem to be associated with a higher risk of all- and high-grade hepatotoxicity compared with control regimens, whereas PD-1 inhibitors seem to be associated with a lower risk of all- and high-grade hepatotoxicity compared with control regimens.
Collapse
Affiliation(s)
- Wenjun Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Puyi Lie
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Minzhang Guo
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jianxing He
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| |
Collapse
|
250
|
Rausch J, Lopez PA, Bialojan A, Denny M, Langguth P, Probst HC, Schild H, Radsak MP. Combined immunotherapy: CTLA-4 blockade potentiates anti-tumor response induced by transcutaneous immunization. J Dermatol Sci 2017; 87:300-306. [PMID: 28666747 DOI: 10.1016/j.jdermsci.2017.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/14/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND The epidermal application of the Toll Like Receptor 7 agonist imiquimod and a T-cell peptide epitope (transcutaneous immunization, TCI) mediates systemic peptide-specific cytotoxic T-cell (CTL) responses and leads to tumor protection in a prophylactic tumor setting. However, it does not accomplish memory formation or permanent defiance of tumors in a therapeutic set-up. As a distinct immunologic approach, CTLA-4 blockade augments systemic immune responses and has shown long-lasting effects in preclinical experiments as well as in clinical trials. OBJECTIVE The study investigates the vaccination capacity of TCI in combination with the checkpoint inhibitor CTLA-4 in matters of primary response, memory formation and tumor protection and characterizes the role of regulatory T cells (Tregs). METHODS After performing TCI with IMI-Sol (containing 5% Imiquimod) and the model epitope SIINFEKL, 6-8 week old C57BL/6 mice received anti-CTLA-4 antibody either s.c or i.p. The CTL responses and frequency of peptide specific CD8+ T-cells were then evaluated on day 8. To determine anti-tumor effects, a therapeutic tumor challenge with B16 OVA melanoma was performed. RESULTS The combination of s.c. anti-CTLA-4 antibody and TCI leads to an enhanced systemic cytotoxic response, to memory formation and allows significantly improved survival in a tumor setting with B16 OVA melanoma. Towards the mechanism, we show that in this vaccination protocol the CTLA-4 antibody acts mainly Treg-independent. CONCLUSION We demonstrate that the combination of TCI with IMI-Sol and anti-CTLA-4 can confer potent immune responses and tumor-protection. These results might contribute to the development of advanced vaccination approaches targeting tumors or persistent infectious diseases.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Aminoquinolines/pharmacology
- Aminoquinolines/therapeutic use
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Drug Synergism
- Flow Cytometry
- Humans
- Imiquimod
- Immunologic Memory/drug effects
- Immunotherapy/methods
- Melanoma, Experimental/immunology
- Melanoma, Experimental/mortality
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/antagonists & inhibitors
- Mice
- Mice, Inbred C57BL
- Ovalbumin/pharmacology
- Ovalbumin/therapeutic use
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Skin Neoplasms/immunology
- Skin Neoplasms/mortality
- Skin Neoplasms/therapy
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Toll-Like Receptor 7/antagonists & inhibitors
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Johanna Rausch
- Third Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Pamela Aranda Lopez
- Third Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ariane Bialojan
- Third Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Mark Denny
- Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Peter Langguth
- Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Markus P Radsak
- Third Department of Medicine - Hematology, Oncology, Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.
| |
Collapse
|