601
|
Freeman M, Betts KA, Jiang S, Du EX, Gupte-Singh K, Lu Y, Rao S, Shoushtari AN. Indirect Treatment Comparison of Nivolumab Versus Observation or Ipilimumab as Adjuvant Therapy in Resected Melanoma Using Pooled Clinical Trial Data. Adv Ther 2019; 36:2783-2796. [PMID: 31440980 PMCID: PMC6822822 DOI: 10.1007/s12325-019-01060-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Nivolumab has been approved in patients with melanoma with lymph node involvement or metastatic disease who have undergone complete resection, in the adjuvant setting. A pivotal trial compared nivolumab with ipilimumab; however, no head-to-head trial exists comparing nivolumab to observation, a common comparator in the adjuvant setting. Here, we compared the efficacy and cost-effectiveness of nivolumab with observation or ipilimumab as adjuvant therapies in resected stage IIIB/C melanoma. METHODS Patient data were pooled from the EORTC 18071 and CheckMate 238 trials using propensity score weighting and adjusting for cross-trial differences. Number needed to treat (NNT) and costs per recurrence-free life-month (RFLM) at 12, 16, 18, and 24 months (as data allowed) were estimated. Costs included drug acquisition, administration costs, and direct medical costs. Sensitivity analyses including patients with stage IIIB/C and resected stage IV melanoma were conducted. RESULTS A total of 1287 patients (278 nivolumab, 365 observation, and 644 ipilimumab) with resected stage IIIB/C melanoma were pooled. NNTs to achieve one additional recurrence-free survivor with nivolumab versus observation were 3.93 at 12 months and 3.42 at 24 months; NNTs for nivolumab versus ipilimumab were 7.97 at 12 months and 6.43 at 24 months. Mean drug costs per RFLM were lower for nivolumab at 12, 18, and 24 months, respectively (nivolumab: $13,447, $9462, and $7370; ipilimumab: $52,734, $40,484, and $33,875). Mean medical costs per RFLM were the lowest for nivolumab versus observation or ipilimumab at 12 months ($449 versus $674 or $1531) and 16 months ($383 versus $808 or $1316). The sensitivity analysis results were consistent with the base case. CONCLUSION For resected melanoma, adjuvant nivolumab is both clinically effective and cost-effective compared with observation or ipilimumab. Adjuvant nivolumab was associated with a lower drug cost per RFLM compared with ipilimumab, and a lower medical cost compared with observation. Future analyses incorporating long-term follow-up data may help increase understanding of the economic impact of nivolumab in the adjuvant setting. FUNDING Bristol-Myers Squibb Company.
Collapse
Affiliation(s)
| | | | - Shan Jiang
- Bristol-Myers Squibb, Princeton, NJ, USA
| | - Ella X Du
- Analysis Group, Inc., Los Angeles, CA, USA
| | | | - Yichen Lu
- Analysis Group, Inc., Los Angeles, CA, USA
| | - Sumati Rao
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|
602
|
Bensimon AG, Zhou ZY, Jenkins M, Song Y, Gao W, Signorovitch J, Krepler C, Liu FX, Wang J, Aguiar-Ibáñez R. Cost-effectiveness of pembrolizumab for the adjuvant treatment of resected high-risk stage III melanoma in the United States. J Med Econ 2019; 22:981-993. [PMID: 31012765 DOI: 10.1080/13696998.2019.1609485] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/27/2019] [Accepted: 04/11/2019] [Indexed: 10/27/2022]
Abstract
Aims: To evaluate the cost-effectiveness of adjuvant pembrolizumab relative to observation alone following complete resection of high-risk stage III melanoma with lymph node involvement, from a US health system perspective. Materials and methods: A Markov cohort model with four health states (recurrence-free, locoregional recurrence, distant metastases, and death) was developed to estimate costs, life-years, and quality-adjusted life-years (QALYs) associated with pembrolizumab vs observation over a lifetime (46-year) horizon. Using a parametric multi-state modeling approach, transition probabilities starting from recurrence-free were estimated based on patient-level data from KEYNOTE-054 (NCT02362594), a direct head-to-head phase 3 trial. Post-recurrence transition probabilities were informed by real-world retrospective data and clinical trials in advanced melanoma. Health state utilities and adverse event-related disutility were derived from KEYNOTE-054 trial data and published literature. Costs of drug acquisition and administration, adverse events, disease management, and terminal care were estimated in 2018 US dollars. Deterministic and probabilistic sensitivity analyses were conducted to assess robustness. Results: Over a lifetime horizon, adjuvant pembrolizumab and observation were associated with total QALYs of 9.24 and 5.95, total life-years of 10.54 and 7.15, and total costs of $489,820 and $440,431, respectively. The resulting incremental cost-effectiveness ratios (ICERs) for pembrolizumab vs observation were $15,009/QALY and $14,550/life-year. Across the range of input values and assumptions tested in deterministic sensitivity analyses, pembrolizumab ranged from being a dominant strategy to having an ICER of $57,449/QALY vs observation. The ICER was below a willingness-to-pay threshold of $100,000/QALY in 90.2% of probabilistic simulations. Limitations: Long-term extrapolation of outcomes was based on interim results from KEYNOTE-054, with a median follow-up of 15 months. Conclusions: Based on common willingness-to-pay benchmarks, pembrolizumab is highly cost-effective compared with observation alone for the adjuvant treatment of completely resected stage III melanoma in the US.
Collapse
Affiliation(s)
| | | | | | - Yan Song
- Analysis Group Inc. , Boston , MA , USA
| | - Wei Gao
- Analysis Group Inc. , Boston , MA , USA
| | | | | | | | | | | |
Collapse
|
603
|
Russell‐Jones R. A brighter future for melanoma. Br J Dermatol 2019; 181:874. [DOI: 10.1111/bjd.18122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
604
|
Egger ME, Scoggins CR, McMasters KM. The Sunbelt Melanoma Trial. Ann Surg Oncol 2019; 27:28-34. [PMID: 31529312 DOI: 10.1245/s10434-019-07828-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Indexed: 12/11/2022]
Abstract
The Sunbelt Melanoma Trial, a multicenter, prospective randomized clinical study, evaluated the role of high-dose interferon alfa-2b (HDI) therapy for patients with a single positive sentinel lymph node (SLN) metastasis treated with a completion lymph node dissection (CLND). A second protocol in the trial evaluated the prognostic significance of using molecular markers to identify submicroscopic metastases in sentinel lymph nodes that were negative by routine pathologic analysis. The role of CLND with or without adjuvant HDI was evaluated in this group of patients. The results of the study demonstrated that adjuvant HDI offered no survival benefit for patients with a single positive SLN in terms of disease-free or overall survival. Molecular staging using polymerase chain reaction (PCR) for melanoma markers did not identify a high-risk group of patients at increased risk of melanoma recurrence. Additional treatment of these patients who were PCR-positive with either CLND alone or CLND plus HDI did not improve their survival. Additional studies from the Sunbelt Melanoma Trial helped to validate the operational standards of the SLN biopsy procedure and defined the complication rates for both SLN biopsy and CLND. A prognostic risk calculator has been developed from trial data, and the importance of different micrometastatic tumor burden measurements was reported. Although the Sunbelt Melanoma Trial did not demonstrate an improvement in survival with HDI, it is an important trial that highlights the significance of surgeon-initiated randomized clinical trials that incorporate surgical techniques, molecular biomarkers, and adjuvant therapy.
Collapse
Affiliation(s)
- Michael E Egger
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Charles R Scoggins
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kelly M McMasters
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
605
|
Bello DM, Faries MB. The Landmark Series: MSLT-1, MSLT-2 and DeCOG (Management of Lymph Nodes). Ann Surg Oncol 2019; 27:15-21. [PMID: 31535299 DOI: 10.1245/s10434-019-07830-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Indexed: 12/30/2022]
Abstract
Management of regional lymph nodes in patients with melanoma has evolved significantly in recent years. The value of nodal intervention, long utilized for its perceived therapeutic benefit, has now shifted to that of a critical prognostic procedure used to guide clinical decision making. This review focuses on the three landmark, randomized controlled trials evaluating the role of surgery for regional lymph nodes in melanoma: Multicenter Selective Lymphadenectomy Trial I (MSLT-I), German Dermatologic Cooperative Oncology Group-Selective Lymphadenectomy Trial (DeCOG-SLT), and Multicenter Selective Lymphadenectomy Trial II (MSLT-II).
Collapse
Affiliation(s)
- Danielle M Bello
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Mark B Faries
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
606
|
Kassi E, Angelousi A, Asonitis N, Diamantopoulos P, Anastasopoulou A, Papaxoinis G, Kokkinos M, Giovanopoulos I, Kyriakakis G, Petychaki F, Savelli A, Benopoulou O, Gogas H. Endocrine-related adverse events associated with immune-checkpoint inhibitors in patients with melanoma. Cancer Med 2019; 8:6585-6594. [PMID: 31518074 PMCID: PMC6825974 DOI: 10.1002/cam4.2533] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background Immune‐checkpoint inhibitors have been shown to improve survival in melanoma patients, but can also trigger immune‐related endocrinopathies, especially hypophysitis and thyroid dysfunction. Methods To assess the incidence and the spectrum of endocrinopathies in melanoma patients treated with immunotherapy a prospective observational study was conducted. Forty out of 339 patients, treated with immune‐checkpoint inhibitors, developed endocrinopathies. All patients had hormonal functional tests at screening (before the initiation of immunotherapy) and during follow‐up. Results The total incidence of endocrinopathies was 11.8%, 13.4% due to anti‐PD1/PDL1, 5% due to anti‐CTLA4, and 18.5% due to sequential and/or combination treatment. Twenty‐one patients (6.2%) presented with isolated anterior hypophysitis, eleven (3.2%) with primary thyroid dysfunction and eight (2.4%) with both abnormalities. The most frequent anterior pituitary hormone deficiency was central adrenal insufficiency, followed by central hypothyroidism and hypogonadotrophic hypogonadism. None of the patients with corticotroph axis failure recovered during follow‐up. Endocrinopathies occurred after a median of 22 weeks (range: 4‐156) from treatment initiation. Of note, sequential and/or combination therapy with anti‐CTLA4 and anti‐PD1/anti‐PDL1 led to an almost threefold incidence of hypophysitis compared to either monotherapy. Only one of 120 patients receiving anti‐CTLA4 monotherapy developed primary hypothyroidism. Conclusions Our cohort demonstrated an increased incidence of hypophysitis with anti‐PD1/anti‐PDL1 in contrast to the rarity of primary thyroid dysfunction with anti‐CTLA4 treatment. These results could be attributed to genetic/ethnic differences. Sequential treatment is, for the first time to our knowledge, reported to increase the risk of developing hypophysitis to a level as high as that of combination therapy.
Collapse
Affiliation(s)
- Eva Kassi
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Angelousi
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Asonitis
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Diamantopoulos
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George Papaxoinis
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Michalis Kokkinos
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilias Giovanopoulos
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Kyriakakis
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Fotini Petychaki
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Akrivi Savelli
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Olga Benopoulou
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Helen Gogas
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
607
|
Parakh S, King D, Gan HK, Scott AM. Current Development of Monoclonal Antibodies in Cancer Therapy. Recent Results Cancer Res 2019; 214:1-70. [PMID: 31473848 DOI: 10.1007/978-3-030-23765-3_1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exploiting the unique specificity of monoclonal antibodies has revolutionized the treatment and diagnosis of haematological and solid organ malignancies; bringing benefit to millions of patients over the past decades. Recent achievements include conjugating antibodies with toxic payloads resulting in superior efficacy and/or reduced toxicity, development of molecular imaging techniques targeting specific antigens for use as predictive and prognostic biomarkers, the development of novel bi- and tri-specific antibodies to enhance therapeutic benefit and abrogate resistance and the success of immunotherapy agents. In this chapter, we review an overview of antibody structure and function relevant to cancer therapy and provide an overview of pivotal clinical trials which have led to regulatory approval of monoclonal antibodies in cancer treatment. We further discuss resistance mechanisms and the unique side effects of each class of antibody and provide an overview of emerging therapeutic agents.
Collapse
Affiliation(s)
- Sagun Parakh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Dylan King
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia.,Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Melbourne, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Melbourne, Australia. .,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia. .,Department of Medicine, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
608
|
The Risk of Diarrhea and Colitis in Patients With Advanced Melanoma Undergoing Immune Checkpoint Inhibitor Therapy: A Systematic Review and Meta-Analysis. J Immunother 2019; 41:101-108. [PMID: 29401166 DOI: 10.1097/cji.0000000000000213] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Checkpoint inhibitors are a first-line therapy for advanced melanoma, though their use is limited by diarrhea and colitis. The aim of our study was to determine the risk of these toxicities associated with immunotherapy in advanced melanoma. Electronic databases were searched through June 2017 for prospective studies reporting the risk of diarrhea and colitis in advanced melanoma treated with anti-programmed death-1 (PD-1) or anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors. Standardized definitions assessed the grade of diarrhea and colitis. Pooled incidence and weighted relative risk estimates with 95% confidence intervals (CI) were estimated using random effects model. Eighteen studies were included: 6 studies (1537 patients) with PD-1 inhibitors and 15 studies (3116 patients) with CTLA-4 inhibitors. The incidence of all-grade diarrhea was 13.7% (95% CI, 10.1%-17.2%) for anti-PD-1 and 35.4% (95% CI, 30.4%-40.5%) for anti-CTLA-4. The incidence of all-grade colitis was 1.6% (95% CI, 0.7%-2.4%) for anti-PD-1, and 8.8% (95% CI, 6.1%-11.5%) for anti-CTLA-4. When PD-1 inhibitors were compared directly with CTLA-4 inhibitors, the relative risk of all-grade diarrhea was 0.58 (95% CI, 0.43-0.77), and the relative risk of all-grade colitis was 0.16 (95% CI, 0.05-0.51). The rate of therapy discontinuation was numerically higher for anti-CTLA-4 therapy compared with anti-PD-1 therapy. Finally, 2 studies compared combination immunotherapy with anti-CTLA-4 therapy alone. The relative risk of developing all-grade diarrhea and colitis with combination therapy was 1.31 (95% CI, 1.09-1.57) and 1.21 (95% CI, 0.73-1.99), respectively. Diarrhea and colitis are frequent toxicities associated with checkpoint inhibitors, and seem to be most common with CTLA-4 inhibitors.
Collapse
|
609
|
Nishida T, Iijima H, Adachi S. Immune checkpoint inhibitor-induced diarrhea/colitis: Endoscopic and pathologic findings. World J Gastrointest Pathophysiol 2019; 10:17-28. [PMID: 31559106 PMCID: PMC6751508 DOI: 10.4291/wjgp.v10.i2.17] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/30/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
The indications of immune checkpoint inhibitors (ICPIs) for cancer treatment have rapidly expanded, and their use is increasing in clinical settings worldwide. Despite the considerable clinical benefits of ICPIs, frequent immune-related adverse events (irAEs) have become nonnegligible concerns. Among irAEs, ICPI-induced colitis/diarrhea is frequent and recognized not only by oncologists but also by gastroenterologists or endoscopists. The endoscopic findings show similarity to those of inflammatory bowel disease to a certain extent, particularly ulcerative colitis, but do not seem to be identical. The pathological findings of ICPI-induced colitis may vary among drug classes. They show acute or chronic inflammation, but it may depend on the time of colitis suggested by colonoscopy, including biopsy or treatment intervention. In the case of chronic inflammation determined by biopsy, the endoscopy findings may overlap with those of inflammatory bowel disease. Here, we provide a comprehensive review of ICPI-induced colitis based on clinical, endoscopic and pathologic findings.
Collapse
Affiliation(s)
- Tsutomu Nishida
- Department of Gastroenterology, Toyonaka Municipal Hospital, Osaka 560-8565, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shiro Adachi
- Department of Pathology, Toyonaka Municipal Hospital, Osaka 560-8565, Japan
| |
Collapse
|
610
|
Roszik J, Markovits E, Dobosz P, Layani A, Slabodnik-Kaner K, Baruch EN, Ben-Betzalel G, Grimm E, Berger R, Sidi Y, Schachter J, Shapira-Frommer R, Avni D, Markel G, Leibowitz-Amit R. TNFSF4 (OX40L) expression and survival in locally advanced and metastatic melanoma. Cancer Immunol Immunother 2019; 68:1493-1500. [PMID: 31501955 DOI: 10.1007/s00262-019-02382-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/20/2019] [Indexed: 01/16/2023]
Abstract
Immunotherapy with checkpoint inhibitors revolutionized melanoma treatment in both the adjuvant and metastatic setting, yet not all metastatic patients respond, and metastatic disease still often recurs among immunotherapy-treated patients with locally advanced disease. TNFSF4 is a co-stimulatory checkpoint protein expressed by several types of immune and non-immune cells, and was shown in the past to enhance the anti-neoplastic activity of T cells. Here, we assessed its expression in melanoma and its association with outcome in locally advanced and metastatic disease. We used publicly available data from The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE), and RNA sequencing data from anti-PD1-treated patients at Sheba medical center. TNFSF4 mRNA is expressed in melanoma cell lines and melanoma samples, including those with low lymphocytic infiltrates, and is not associated with the ulceration status of the primary tumor. Low expression of TNFSF4 mRNA is associated with worse prognosis in all melanoma patients and in the cohorts of stage III and stage IIIc-IV patients. Low expression of TNFSF4 mRNAs is also associated with worse prognosis in the subgroup of patients with low lymphocytic infiltrates, suggesting that tumoral TNFSF4 is associated with outcome. TNFSF4 expression was not correlated with the expression of other known checkpoint mRNAs. Last, metastatic patients with TNFSF4 mRNA expression within the lowest quartile have significantly worse outcome on anti-PD1 treatment, and a significantly lower response rate to these agents. Our current work points to TNFSF4 expression in melanoma as a potential determinant of prognosis, and warrants further translational and clinical research.
Collapse
Affiliation(s)
- Jason Roszik
- Departments of Melanoma Medical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Ettai Markovits
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Paula Dobosz
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Adi Layani
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Keren Slabodnik-Kaner
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Erez N Baruch
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Guy Ben-Betzalel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Elizabeth Grimm
- Departments of Melanoma Medical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Raanan Berger
- Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.,Division of Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Oncology Institute and Cancer Research Center, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Yehezkel Sidi
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.,Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Ronnie Shapira-Frommer
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Dror Avni
- Lab of Molecular Cancer Research, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel. .,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Raya Leibowitz-Amit
- Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel. .,Division of Oncology, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel. .,Oncology Institute and Cancer Research Center, Sheba Medical Center-Tel Hashomer, 2 Sheba Road, 5266202, Ramat Gan, Israel.
| |
Collapse
|
611
|
Verver D, Poirier-Colame V, Tomasic G, Cherif-Rebai K, Grunhagen DJ, Verhoef C, Suciu S, Robert C, Zitvogel L, Eggermont AMM. Upregulation of intratumoral HLA class I and peritumoral Mx1 in ulcerated melanomas. Oncoimmunology 2019; 8:e1660121. [PMID: 31646109 DOI: 10.1080/2162402x.2019.1660121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/22/2019] [Indexed: 12/27/2022] Open
Abstract
Before the era of immune checkpoint blockade, a meta-analysis encompassing fifteen trials reported that adjuvant IFN-α significantly reduces the risk of relapse and improves survival of ulcerated melanoma (UM) with no benefit for higher doses compared to lower doses. IFNa2b affects many cell intrinsic features of tumor cells and modulates the host innate and cognate immune responses. To better understand the biological traits associated with ulceration that could explain the efficacy of prophylactic type 1 IFN, we performed immunohistochemical analysis of various molecules (major histocompatibility complex class I and class II, MX Dynamin Like GTPase 1 (MX1), inducible Nitric-Oxide Synthase (iNOS) or CD47) in two retrospective cohorts of melanoma patients, one diagnosed with a primary cutaneous melanoma (1995-2013, N = 172, among whom 49% were ulcerated melanoma (UM)) and a second one diagnosed with metastatic melanoma amenable to lymph node resection (EORTC 18952 and 18991 trials, N = 98, among whom 44% were UM). We found that primary and metastatic UM exhibit higher basal expression of MHC class I molecules, independently of Breslow thickness, histology and lymphocytic infiltration compared with NUM and that primary UM harbored higher constitutive levels of the antiviral protein Mx1 at the border of tumor beds than NUM. These findings suggest that UM expand in a tumor microenvironment where chronic exposure to type 1 IFN could favor a response to exogenous IFNs.
Collapse
Affiliation(s)
- Daniëlle Verver
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Vichnou Poirier-Colame
- Department of Immuno-Oncology, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Gorana Tomasic
- Department of Pathology, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Khadija Cherif-Rebai
- Department of Pathology, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Dirk J Grunhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Stefan Suciu
- Department of Biostatistics, European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Caroline Robert
- Department of Medicine, Service of Dermatology Gustave Roussy and University Paris-Sud
| | - Laurence Zitvogel
- INSERM U 1015, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Alexander M M Eggermont
- INSERM U 1015, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France.,University Paris-Sud, Le Kremlin Bicetre, France
| |
Collapse
|
612
|
Song Y, Azari FS, Metzger DA, Fraker DL, Karakousis GC. Practice Patterns and Prognostic Value of Sentinel Lymph Node Biopsy for Thick Melanoma: A National Cancer Database Study. Ann Surg Oncol 2019; 26:4651-4662. [PMID: 31485823 DOI: 10.1245/s10434-019-07783-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Sentinel lymph node biopsy (SLNB) has been somewhat controversial for patients with a diagnosis of thick (> 4 mm) melanoma. This study aimed to characterize the national practice pattern in performing SLNB for this patient population and to determine the predictors and prognostic value of nodal positivity using population-level data. METHODS Patients with a diagnosis of clinically node-negative, thick melanoma (2010-2015) were identified using the National Cancer Database. Factors associated with performing regional nodal evaluation were characterized. Predictors of nodal positivity were determined using multivariable logistic regression. Overall survival (OS) was estimated using standard statistical methods. RESULTS Of 9847 study patients, 7513 (76.3%) underwent SLNB. The patients who underwent nodal evaluation were younger (median age, 66 vs 81 years; P < 0.001), less likely to have comorbid conditions (19.6% vs 26.0%; P < 0.001), more often privately insured (40.4% vs 16.4%; P < 0.001), and more frequently treated at an academic center (49.5% vs 43.9%; P < 0.001). Among those who underwent nodal evaluation, 25.5% had metastatic nodes. Multivariable regression identified age, Charlson-Deyo score, primary location, ulceration, mitoses, vertical growth phase, and lymphovascular invasion as independent predictors of nodal positivity, but with only moderate predictive accuracy (optimism-adjusted area under the curve, 0.684). Furthermore, compared with node negativity, node positivity was significantly associated with decreased OS (hazard ratio, 2.05; P < 0.001). CONCLUSION Although nodal status provides important prognostic information, at a national level, nearly one fourth of patients with clinically node-negative, thick melanoma do not undergo SLNB. Appropriate pathologic staging would allow these high-risk patients to be candidates for effective adjuvant therapy.
Collapse
Affiliation(s)
- Yun Song
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Feredun S Azari
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Aryeh Metzger
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas L Fraker
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
613
|
Abstract
Immune checkpoint blockers have revolutionized cancer treatment in recent years. These agents are now approved for the treatment of several malignancies, including melanoma, squamous and non-squamous non-small cell lung cancer, renal cell carcinoma, urothelial carcinoma, and head and neck squamous cell carcinoma. Studies have demonstrated the significant impact of immunotherapy versus standard of care on patient outcomes, including durable response and extended survival. The use of immunotherapy-based combination therapy has been shown to further extend duration of response and survival. Immunotherapies function through modulation of the immune system, which can lead to immune-mediated adverse events (imAEs). These include a range of dermatologic, gastrointestinal, endocrine, and hepatic toxicities, as well as other less common inflammatory events. ImAEs are typically low grade and manageable when identified early and treated with appropriate measures. Identifying the right patient for the right therapy will become more important as new immunotherapies and immunotherapy-based combinations are approved and costs of cancer care continue to rise.
Collapse
|
614
|
Isaksson K, Katsarelias D, Mikiver R, Carneiro A, Ny L, Olofsson Bagge R. A Population-Based Comparison of the AJCC 7th and AJCC 8th Editions for Patients Diagnosed with Stage III Cutaneous Malignant Melanoma in Sweden. Ann Surg Oncol 2019; 26:2839-2845. [PMID: 31111349 PMCID: PMC6682854 DOI: 10.1245/s10434-019-07448-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cutaneous melanoma is steadily increasing worldwide. The new AJCC 8th edition was recently launched and introduced several changes in melanoma staging, particularly for stage III. We conducted a population-based registry study with the purpose to evaluate the impact and prognostic accuracy of the new classification in Sweden. METHODS Consecutive patients diagnosed with stage III melanoma between January 2005 and September 2017 were identified by the Swedish Melanoma Registry (SMR) and included for analyses. Patients with multiple primary melanomas were excluded. Patients were classified according to the AJCC 7th as well as the 8th edition. Melanoma-specific survival (MSS) was retrieved from the Swedish Cause of Death Registry. RESULTS A total of 2067 eligible patients were identified from the SMR; 1150 patients (57%) changed stage III subgroup when reclassified according to the AJCC 8th edition. The median 5- and 10-year MSS for the whole cohort of stage III melanoma patients was 59% and 51% respectively. The MSS for substage IIIA, B, and C were all improved when patients were reclassified by using to the AJCC 8th edition. The newly defined substage IIID had the worst prognosis with a 10-year MSS of 16%. CONCLUSIONS A high proportion of patients diagnosed with stage III melanoma in Sweden between 2005 and 2017 was restaged to another subgroup, when they were reclassified according to the AJCC 8th of staging manual. We established an improved MSS for all substages compared with the former AJCC 7th edition. This may have implications on decisions about adjuvant treatment.
Collapse
Affiliation(s)
- Karolin Isaksson
- Department of Clinical Sciences Lund, Surgery, Lund University, Skåne University Hospital, Lund, Sweden.
| | - Dimitrios Katsarelias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rasmus Mikiver
- Department of Clinical and Experimental Medicine, Regional Cancer Center South East Sweden, Linköping University, Linköping, Sweden
| | - Ana Carneiro
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
615
|
Hughes MS, Zheng H, Zubiri L, Molina GE, Chen ST, Mooradian MJ, Allen IM, Reynolds KL, Dougan M. Colitis after checkpoint blockade: A retrospective cohort study of melanoma patients requiring admission for symptom control. Cancer Med 2019; 8:4986-4999. [PMID: 31286682 PMCID: PMC6718531 DOI: 10.1002/cam4.2397] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (CPIs) have revolutionized oncologic therapy but can lead to immune-related adverse events (irAEs). Corticosteroids are first-line treatment with escalation to biologic immunosuppression in refractory cases. CPI-related gastroenterocolitis (GEC) affects 20%-50% of patients receiving CPIs and can carry significant morbidity and mortality. Severe CPI-related GEC is not well-described. We present the clinical characterization of all CPI-related GEC requiring admission at a single institution. METHODS Clinical, laboratory, radiographic, and endoscopic data were extracted from charts of all melanoma patients ≥18 years of age admitted to one institution for CPI-related GEC, from February 5, 2011 to December 13, 2016. Patients were followed until December 31, 2017 for further admissions. Survival, outcomes, and pharmaceutical-use analyses were performed. RESULTS Median time-to-admission from initial CPI exposure was 73.5 days. Median length of stay was 4.5 days. About 50.0% required second-line immunosuppression. Readmission for recrudescence occurred in 33.3%. Common Terminology Criteria for Adverse Events (CTCAE) grade was not significantly associated with outcomes. Hypoalbuminemia (P = 0.005), relative lymphopenia (P = 0.027), and decreased lactate dehydrogenase (P = 0.026) were associated with second-line immunosuppression. There was no difference in progression-free survival (PFS) or OS (P = 0.367, 0.400) for second-line immunosuppression. Subgroup analysis showed that early corticosteroid administration (P = 0.045) was associated with decreased PFS. CONCLUSIONS Severe CPI-related GEC typically manifests within 3 months of immunotherapy exposure. Rates of second-line immunosuppression and readmission for recrudescence were high. CTCAE grade did not capture the degree of severity in our cohort. Second-line immunosuppression was not associated with poorer oncologic outcomes; however, early corticosteroid exposure was associated with decreased PFS. Further investigation is warranted.
Collapse
Affiliation(s)
- Michael S. Hughes
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
| | - Hui Zheng
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
| | - Leyre Zubiri
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
| | - Gabriel E. Molina
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
| | - Steven T. Chen
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
- Department of DermatologyMassachusetts General HospitalBostonMassachusetts
| | - Meghan J. Mooradian
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
- Massachusetts General Hospital Cancer CenterBostonMassachusetts
| | - Ian M. Allen
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
| | - Kerry L. Reynolds
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
- Massachusetts General Hospital Cancer CenterBostonMassachusetts
| | - Michael Dougan
- Harvard Medical SchoolBostonMassachusetts
- Department of MedicineMassachusetts General HospitalBostonMassachusetts
- Division of GastroenterologyMassachusetts General HospitalBostonMassachusetts
| |
Collapse
|
616
|
|
617
|
Eggermont AM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, Schmidt H, Hamid O, Robert C, Ascierto PA, Richards JM, Lebbe C, Ferraresi V, Smylie M, Weber JS, Maio M, Hosein F, de Pril V, Kicinski M, Suciu S, Testori A. Adjuvant ipilimumab versus placebo after complete resection of stage III melanoma: long-term follow-up results of the European Organisation for Research and Treatment of Cancer 18071 double-blind phase 3 randomised trial. Eur J Cancer 2019; 119:1-10. [DOI: 10.1016/j.ejca.2019.07.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 12/19/2022]
|
618
|
Simonaggio A, Michot JM, Voisin AL, Le Pavec J, Collins M, Lallart A, Cengizalp G, Vozy A, Laparra A, Varga A, Hollebecque A, Champiat S, Marabelle A, Massard C, Lambotte O. Evaluation of Readministration of Immune Checkpoint Inhibitors After Immune-Related Adverse Events in Patients With Cancer. JAMA Oncol 2019; 5:1310-1317. [PMID: 31169866 PMCID: PMC6555478 DOI: 10.1001/jamaoncol.2019.1022] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Although immune checkpoint inhibitors (ICIs), such as anti-PD-1 (programmed cell death 1) or anti-PD-L1 (programmed cell death 1 ligand 1), have proved effective in treating many cancers, patients receiving ICIs may experience immune-related adverse events (irAEs). Little evidence exists on the safety of resuming these treatments after an irAE. OBJECTIVE To investigate the safety of a rechallenge with anti-PD-1 or anti-PD-L1 immunotherapies after an irAE. DESIGN, SETTING, AND PARTICIPANTS This cohort study of the safety of an ICI rechallenge involved consecutive adult patients (n = 93) who were referred to the ImmunoTOX assessment board at the Gustave Roussy cancer center in Villejuif, France, between August 1, 2015, and December 31, 2017. Data were analyzed from May 28 to November 25, 2018. MAIN OUTCOMES AND MEASURES Incidence of a second irAE in patients who had a readministration of an anti-PD-1 or anti-PD-L1 inhibitor after an initial grade 2 or higher irAE. Characteristics of the patients and the irAEs were reviewed, and the primary end point was the rate of occurrence of second irAEs. RESULTS A total of 93 patients were included, among whom 48 (52%) were female, and the median (range) age was 62.5 (33-85) years. The main cancer types or tumor sites were melanoma (31 [33%]), lung (15 [16%]), colorectal (8 [9%]), and lymphoma (8 [9%]). For the initial irAE, 43 grade 2 events (46%), 36 grade 3 events (39%), and 14 grade 4 events (15%) were found, presenting primarily as hepatitis (17 [18%]), skin toxic effect (14 [15%]), pneumonitis (13 [14%]), colitis (11 [12%]), or arthralgia (7 [7.5%]). Forty patients (43%) were rechallenged with the same anti-PD-1 or anti-PD-L1 agent. The rechallenged and non-rechallenged groups did not differ in terms of median (range) age (61 [34-84] years vs 63 [33-85] years; P = .37), time to initial irAE (5 [1-40] treatment cycles vs 3 [1-22] treatment cycles; P = .32), irAE severity (grade 2: 18 [47.5%] vs 27 [51%]; grades 3-4: 22 [52.5%] vs 26 [49%]; P = .70), or steroid use (17 [42.5%] vs 32 [60%]; P = .09). With a median follow-up period of 14 months, the same irAE or a different irAE occurred in 22 patients (55%). Shorter time to the initial irAE was linked to the occurrence of a second irAE (9 vs 15 weeks; P = .04). The second irAEs were not found to be more severe than the first. CONCLUSIONS AND RELEVANCE The risk-reward ratio for an anti-PD-1 or anti-PD-L1 rechallenge appears to be acceptable, although these patients require close monitoring; further investigation into rechallenge conditions through a prospective clinical trial is needed.
Collapse
Affiliation(s)
- Audrey Simonaggio
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Jean Marie Michot
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
- Department of Internal Medicine and Clinical Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Bicêtre, Le Kremlin-Bicêtre, France
| | | | - Jérome Le Pavec
- Department of Thoracic and Cardiovascular Surgery and Heart and Lung Transplantation, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Michael Collins
- Gastroenterology Unit, Université Paris-Sud, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Bicêtre, Le Kremlin-Bicêtre, France
- Inserm U1193, Paul-Brousse University Hospital, Hepatobiliary Center, Villejuif, France
| | - Audrey Lallart
- Pharmacovigilance Unit, Gustave Roussy, Villejuif, France
| | | | - Aurore Vozy
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Ariane Laparra
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Andréa Varga
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | | | - Stéphane Champiat
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Aurélien Marabelle
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Christophe Massard
- Department of Drug Development (DITEP), Gustave Roussy, Villejuif, France
| | - Olivier Lambotte
- Department of Internal Medicine and Clinical Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Bicêtre, Le Kremlin-Bicêtre, France
- Inserm U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- Université Paris-Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, Infectious Disease Models and Innovative Therapies, Fontenay-aux-Roses, France
| |
Collapse
|
619
|
Weiss SA, Wolchok JD, Sznol M. Immunotherapy of Melanoma: Facts and Hopes. Clin Cancer Res 2019; 25:5191-5201. [PMID: 30923036 PMCID: PMC6726509 DOI: 10.1158/1078-0432.ccr-18-1550] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Melanoma is among the most sensitive of malignancies to immune modulation. Although multiple trials conducted over decades with vaccines, cytokines, and cell therapies demonstrated meaningful responses in a small subset of patients with metastatic disease, a true increase in overall survival (OS) within a randomized phase III trial was not observed until the development of anti-CTLA-4 (ipilimumab). Further improvements in OS for metastatic disease were observed with the anti-PD-1-based therapies (nivolumab, pembrolizumab) as single agents or combined with ipilimumab. A lower bound for expected 5-year survival for metastatic melanoma is currently approximately 35% and could be as high as 50% for the nivolumab/ipilimumab combination among patients who would meet criteria for clinical trials. Moreover, a substantial fraction of long-term survivors will likely remain progression-free without continued treatment. The hope and major challenge for the future is to understand the immunobiology of tumors with primary or acquired resistance to anti-PD-1 or anti-PD-1/anti-CTLA-4 and to develop effective immune therapies tailored to individual patient subsets not achieving long-term clinical benefit. Additional goals include optimal integration of immune therapy with nonimmune therapies, the development and validation of predictive biomarkers in the metastatic setting, improved prognostic and predictive biomarkers for the adjuvant setting, understanding mechanisms of and decreasing toxicity, and optimizing the duration of therapy.
Collapse
Affiliation(s)
- Sarah A Weiss
- Yale University School of Medicine, New Haven, Connecticut.
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mario Sznol
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
620
|
Katsarelias D, Isaksson K, Olofsson Bagge R. ASO Author Reflections: Effects of the Updated AJCC Classification for Patients with Stage III Melanoma. Ann Surg Oncol 2019; 26:713-714. [PMID: 31468215 DOI: 10.1245/s10434-019-07765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 11/18/2022]
Affiliation(s)
- Dimitrios Katsarelias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences Lund, Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden. .,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
621
|
Ollila DW, Meyers MO. Time may Heal All Wounds, but While It Does, Melanoma Marches on. Ann Surg Oncol 2019; 26:3800-3802. [PMID: 31468216 DOI: 10.1245/s10434-019-07674-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Indexed: 11/18/2022]
Affiliation(s)
- David W Ollila
- Department of Surgery, Division of Surgical Oncology, University of North Carolina, Chapel Hill, NC, USA.
| | - Michael O Meyers
- Department of Surgery, Division of Surgical Oncology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
622
|
Abstract
Immunomodulatory antibodies that directly trigger and reawaken suppressed T-cell effector function are termed 'checkpoint inhibitors'. CTLA-4 and PD-1/PD-L1 molecules are the most studied inhibitory immune check points against cancer and because of this therapeutic property have entered the clinic for treating a variety of tumor types. The results so far demonstrate a positive impact on cancer remission. Preclinical studies have demonstrated that targeting a number of other T-cell surface molecules including both positive and negative immune regulators, also possesses strong antitumor activity. Some of these molecules have already entered clinical trials. In this report, we briefly highlight the status of these immune checkpoint inhibitors and discuss their side effects and future directions for their use.
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA.,Eutilex Institute for Biomedical Research, Suite #1401 Daeryung Technotown 17, Gasan digital 1-ro 25, Geumcheon-gu, Seoul Korea
| |
Collapse
|
623
|
Pierantoni F, Maruzzo M, Gardi M, Bezzon E, Gardiman MP, Porreca A, Basso U, Zagonel V. Immunotherapy and urothelial carcinoma: An overview and future prospectives. Crit Rev Oncol Hematol 2019; 143:46-55. [PMID: 31476551 DOI: 10.1016/j.critrevonc.2019.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Urothelial carcinoma (UC) is a common malignancy with a high mortality rate when metastatic. Traditionally, systemic therapy consisted in platinum-based regimens as first-line, with Taxanes or Vinflunine as further lines. Recently, checkpoint inhibitors (CPIs) immunotherapy has emerged as a new therapeutic option. METHODS We searched in Medline, Pubmed and ClinicalTrial.gov databases for the relevant literature, reviewing the results of published trials and the design of ongoing studies involving CPIs in UC. RESULT Strong evidence supports the use of CPIs after failure of Cisplatin-based chemotherapy, although no predictive parameter is available so far. Expression of Programmed-Death-1-Ligand has given conflicting results, and is currently indicated only for the selection of Cisplatin-ineligible patients who should receive CPIs. CONCLUSION The therapeutic landscape of UC is rapidly changing due to the availability of CPIs. Neoadjuvant trials with CPIs and trials combining two CPIs are promising and will further expand the use of immunotherapy.
Collapse
Affiliation(s)
- Francesco Pierantoni
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy.
| | - Marco Maruzzo
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Mario Gardi
- Urology Unit, Department of Surgery, Sant'Antonio Hospital, Padua, Italy
| | - Elisabetta Bezzon
- Radiology Unit, Department of Imaging and Medical Physics, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Marina Paola Gardiman
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University Hospital of Padua, Italy
| | - Angelo Porreca
- Urology Unit, Policlinico Abano Terme, Abano Terme, Italy
| | - Umberto Basso
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| | - Vittorina Zagonel
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCSS, Padua, Italy
| |
Collapse
|
624
|
Spagnolo F, Boutros A, Tanda E, Queirolo P. The adjuvant treatment revolution for high-risk melanoma patients. Semin Cancer Biol 2019; 59:283-289. [PMID: 31445219 DOI: 10.1016/j.semcancer.2019.08.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/05/2019] [Accepted: 08/20/2019] [Indexed: 01/06/2023]
Abstract
The past 5 years have witnessed the results of many practice-changing studies that have dramatically improved the landscape of adjuvant therapy in patients with resected, high-risk melanoma. After a 20-year era of adjuvant interferon, the anti-CTLA-4 and anti-PD-1 immune-checkpoint inhibitors, and MAPK-directed targeted therapy brought a revolution into the adjuvant treatment of melanoma. These results came along with the practice-changing results of two large multicenter studies showing no benefit in terms of overall survival for completion lymph node dissection after positive sentinel node biopsy. In this review, we summarized the current state of the art of the adjuvant treatment of high-risk melanoma, with a view on future perspectives.
Collapse
Affiliation(s)
| | - Andrea Boutros
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrica Tanda
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
625
|
New paradigm for stage III melanoma: from surgery to adjuvant treatment. J Transl Med 2019; 17:266. [PMID: 31412885 PMCID: PMC6693227 DOI: 10.1186/s12967-019-2012-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/03/2019] [Indexed: 12/19/2022] Open
Abstract
Background Recently the 8th version of the American Joint Committee on Cancer (AJCC) classification has been introduced, and has attempted to define a more accurate and precise definition of prognosis in line with the major progresses in understanding the biology and pathogenesis of melanoma. This new staging system introduces major changes in the stage III staging system. Indeed, surgical practice is changing in stage III patients, since, according to recent evidence, there is no survival benefit in radical lymph node dissection following a positive sentinel lymph node dissection. Therefore, some patients currently staged IIIB-C after dissection could be downgraded to IIIA (as in the case of patients with metastatic non-sentinel lymph nodes) since many completion lymph node dissections will no longer be performed. Moreover, new and effective targeted and immune strategies are being introduced in the pharmacological armamentarium in the adjuvant setting, showing major efficacy. Conclusions This article provides the authors’ personal view on the above-mentioned topics.
Collapse
|
626
|
Chen X, Chen X, Gao J, Yang H, Duan Y, Feng Y, He X, Gong X, Wang H, Wu X, Chang J. Astragaloside III Enhances Anti-Tumor Response of NK Cells by Elevating NKG2D and IFN-γ. Front Pharmacol 2019; 10:898. [PMID: 31456687 PMCID: PMC6701288 DOI: 10.3389/fphar.2019.00898] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/16/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells play an irreplaceable role in the development of colon cancer, in which antitumor function of NK cells was impaired. Astragaloside III is a natural compound from Astragalus that has been shown to have immunomodulatory effects in various systems. However, few studies have evaluated the antitumor effects of Astragaloside III through stimulating systemic immunity and regulating NK cells. In this study, flow cytometry, immunohistochemical analysis, and immunofunctional assays were performed to elucidate the functions of Astragaloside III in restoring antitumor function of NK cells. We demonstrated that Astragaloside III significantly elevated the expression of natural killer group 2D (NKG2D), Fas, and interferon-γ (IFN-γ) production in NK cells, leading to increased tumor-killing ability. Experiments in cell co-culture assays and CT26-bearing mice model further confirmed that Astragaloside III could effectively impede tumor growth by increasing infiltration of NK cells into tumor and upregulating the antitumor response of NK cells. We further revealed that Astragaloside III increased IFN-γ secretion of NK cells by enhancing the expression of transcription factor T-bet. In conclusion, the effective anti-tumor function of Astragaloside III was achieved through up-regulation of the immune response of NK cells and elevation of NKG2D, Fas, and IFN-γ production.
Collapse
Affiliation(s)
- Xingmeng Chen
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junxiao Gao
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Han Yang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yue Duan
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yuxin Feng
- Tianjin Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoqun Gong
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Hanjie Wang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoli Wu
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Jin Chang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
627
|
Dubey D, David WS, Amato AA, Reynolds KL, Clement NF, Chute DF, Cohen JV, Lawrence DP, Mooradian MJ, Sullivan RJ, Guidon AC. Varied phenotypes and management of immune checkpoint inhibitor-associated neuropathies. Neurology 2019; 93:e1093-e1103. [PMID: 31405908 DOI: 10.1212/wnl.0000000000008091] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/22/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To describe the spectrum, clinical course, and management of neuropathies associated with immune checkpoint inhibitors (ICIs). METHODS Patients with ICI-related neuropathy (irNeuropathy) were identified and their clinical characteristics compared to neuropathy attributed to cytotoxic agents. RESULTS We identified 19 patients with irNeuropathies. ICIs included anti-programmed death-1 (PD1), 9; anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA4), 2; and combination of anti-CTLA4 and anti-PD1, 8. Median number of ICI doses prior to neuropathy onset was 4. Rate of neuropathies following ICI therapy was 0.7%. Underlying malignancies included melanoma (n = 15), lung adenocarcinoma (n = 3), and cholangiocarcinoma (n = 1). Neuropathy phenotypes were cranial neuropathies with or without meningitis (n = 7), nonlength-dependent polyradiculoneuropathies with and without cranial nerve involvement (n = 6), small-fiber/autonomic neuropathy (n = 2), ANCA-associated mononeuritis multiplex (n = 1), sensory neuronopathy (n = 1), length-dependent sensorimotor axonal polyneuropathy (n = 1), and neuralgic amyotrophy (n = 1). Immune-related adverse events involving other organ systems were common (58%). Corticosteroid use for management of neuropathy was associated with improvement in median modified Rankin Scale score (1 vs 0, p = 0.001) and Inflammatory Neuropathy Cause and Treatment Disability score (2 vs 0.5, p = 0.012) (Class IV). Significantly higher proportion of irNeuropathies had acute or subacute and nonlength-dependent presentations (p < 0.001) and rate of hospitalization for irNeuropathy was also higher (p = 0.002) compared to toxic neuropathy from chemotherapy. CONCLUSION Neuropathy is a rare complication of ICIs that often responds to immunosuppression. Recognition of its wide phenotypic spectrum and distinct clinical characteristics and prompt management with corticosteroids may lead to favorable outcomes.
Collapse
Affiliation(s)
- Divyanshu Dubey
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - William S David
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Anthony A Amato
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Kerry L Reynolds
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Nathan F Clement
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Donald F Chute
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Justine V Cohen
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Donald P Lawrence
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Meghan J Mooradian
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Ryan J Sullivan
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN
| | - Amanda C Guidon
- From the Departments of Neurology (D.D., W.S.D., A.C.G.), Medicine (K.L.R., D.F.C., J.V.C., D.P.L., M.J.M., R.J.S.), and Pathology (N.F.C.), Massachusetts General Hospital; Department of Neurology (D.D., A.A.A.), Brigham and Women's Hospital, Boston, MA; and Department of Neurology (D.D.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
628
|
Abstract
Immunotherapy has dramatically improved the prognosis for patients with melanoma and has become the cornerstone of treatment for those with advanced disease. The role of immunotherapy continues to expand with multiple new agents approved in the adjuvant as well as metastatic setting, as first-line therapy and beyond. We review the currently approved drugs for the treatment of melanoma, along with clinical trial data, adverse side effects, response assessment and future directions.
Collapse
Affiliation(s)
- Emily Feld
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
629
|
Teufel A, Zhan T, Härtel N, Bornschein J, Ebert MP, Schulte N. Management of immune related adverse events induced by immune checkpoint inhibition. Cancer Lett 2019; 456:80-87. [DOI: 10.1016/j.canlet.2019.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/02/2019] [Accepted: 04/09/2019] [Indexed: 12/17/2022]
|
630
|
Khunger A, Buchwald ZS, Lowe M, Khan MK, Delman KA, Tarhini AA. Neoadjuvant therapy of locally/regionally advanced melanoma. Ther Adv Med Oncol 2019; 11:1758835919866959. [PMID: 31391869 PMCID: PMC6669845 DOI: 10.1177/1758835919866959] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/08/2019] [Indexed: 11/18/2022] Open
Abstract
Locally/regionally advanced melanoma confers a major challenge in terms of surgical and medical management. Surgical treatment carries the risks of surgical morbidities and potential complications that could be lasting. In addition, these patients continue to have a high risk of relapse and death despite the use of standard adjuvant therapy. Neoadjuvant therapy has the potential to significantly improve the clinical outcome of these patients, particularly in this era of newer and effective targeted and immunotherapeutic agents. Previous neoadjuvant studies tested chemotherapy with temozolomide where the clinical activity was limited. Biochemotherapy (BCT) was tested in two studies in the neoadjuvant setting and showed high tumor response rates; however, BCT was ultimately abandoned following its failure to demonstrate survival benefits in randomized trials of metastatic disease. Success of immunotherapy and targeted therapy in prolonging the lives of patients with metastatic melanoma generated considerable interest to investigate these novel strategies in the adjuvant and neoadjuvant settings. A number of neoadjuvant targeted and immunotherapy studies have been completed in melanoma to date and have yielded promising clinical activity. Given these encouraging results, a number of studies with other molecularly targeted and immunotherapeutic agents and their combinations are ongoing in the neoadjuvant setting; long-term outcome data are eagerly awaited. Such studies also provide access to biospecimens before and during therapy, allowing for the conduct of biomarker and mechanistic studies that may have a significant impact in guiding adjuvant therapy choices and drug development.
Collapse
Affiliation(s)
- Arjun Khunger
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, USA
| | - Zachary S. Buchwald
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Lowe
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammad K. Khan
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Keith A. Delman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Ahmad A. Tarhini
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Comprehensive Cancer Center, 1365 Clifton Rd Atlanta, GA 30322, USA
| |
Collapse
|
631
|
Pitroda SP, Chmura SJ, Weichselbaum RR. Integration of radiotherapy and immunotherapy for treatment of oligometastases. Lancet Oncol 2019; 20:e434-e442. [PMID: 31364595 DOI: 10.1016/s1470-2045(19)30157-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
Metastasis is the leading cause of cancer-related mortality and remains one of the prevailing challenges in cancer treatment. Most patients with metastatic disease are treated with systemic agents, which prolong survival and improve symptoms but are typically not curative. The oligometastatic hypothesis challenges the perspective that metastasis is an invariably disseminated process, and proposes a biological spectrum of metastatic virulence. Mounting evidence supports the idea that patients with numerically and spatially restricted sites of metastases, termed oligometastases, can achieve prolonged survival following metastasis-directed therapies, such as surgery or radiotherapy. Improvements in clinical and molecular staging of metastatic disease, as well as integration of effective systemic therapies with localised interventions, might achieve better outcomes for patients with diverse metastatic states. In this Series paper, we propose a rationale for the integration of immune checkpoint inhibitors with radiotherapy to advance the potential for effective treatment along the spectrum of disease, with emphasis on how immunotherapy can potentiate radiotherapy treatment in the oligometastatic setting.
Collapse
Affiliation(s)
- Sean P Pitroda
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA; Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Steven J Chmura
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA; Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
632
|
Lorenzi M, Arndorfer S, Aguiar-Ibañez R, Scherrer E, Liu FX, Krepler C. An indirect treatment comparison of the efficacy of pembrolizumab versus competing regimens for the adjuvant treatment of stage III melanoma. J Drug Assess 2019; 8:135-145. [PMID: 31489255 PMCID: PMC6713115 DOI: 10.1080/21556660.2019.1649266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
Objective: To determine the efficacy of pembrolizumab relative to other treatments used in stage III melanoma by conducting a systematic literature review (SLR) and network meta-analysis (NMA). Methods: A SLR was conducted to identify randomized clinical trials (RCTs) evaluating approved adjuvant treatments including interferon-containing regimens, BRAF-inhibitors, and PD-L1 inhibitors in stage III melanoma patients. Relative treatment effects for recurrence-free survival (RFS) were synthesized with Bayesian NMA models that allowed for hazard ratios (HRs) to vary over time. Results: Included studies formed a connected network of evidence composed of eight trials. In high-risk stage III patients, the HR for pembrolizumab vs observation decreased significantly over time with the superiority of pembrolizumab over observation becoming statistically meaningful before 3 months. By 9 months, the HR for pembrolizumab vs observation was statistically significantly lower than the HR for most other treatments vs observation, with the exception of ipilimumab and biochemotherapy due to overlapping 95% credible intervals. In BRAF + patients, pembrolizumab was statistically significantly better than observation after 3 months. The HR for both BRAF-inhibitors vs observation increased significantly over time and pembrolizumab was statistically superior to both BRAF-inhibitors after 15 months. Conclusions: Pembrolizumab results in statistically significantly improved RFS compared to all competing regimens after 9 months, except ipilimumab and biochemotherapy, for the adjuvant treatment of stage III melanoma. However, point estimate HRs vs observation for pembrolizumab are much lower than those for ipilimumab. In BRAF + patients, the advantage of pembrolizumab versus competing interventions increases over time with respect to RFS.
Collapse
|
633
|
Perspectives in melanoma: meeting report from the Melanoma Bridge (November 29th-1 December 1st, 2018, Naples, Italy). J Transl Med 2019; 17:234. [PMID: 31331337 PMCID: PMC6647284 DOI: 10.1186/s12967-019-1979-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/10/2019] [Indexed: 01/07/2023] Open
Abstract
Diagnosis of melanocytic lesions, correct prognostication of patients, selection of appropriate adjuvant and systemic therapies, and prediction of response to a given therapy remain very real challenges in melanoma. Recent studies have shown that immune checkpoint blockade that represents a forefront in cancer therapy, provide responses but they are not universal. Improved understanding of the tumor microenvironment, tumor immunity and response to therapy has prompted extensive translational and clinical research in melanoma. Development of novel biomarker platforms may help to improve diagnostics and predictive accuracy for selection of patients for specific treatment. There is a growing evidence that genomic and immune features of pre-treatment tumor biopsies may correlate with response in patients with melanoma and other cancers they have yet to be fully characterized and implemented clinically. For example, advancements in sequencing and the understanding of the tumor microenvironment in melanoma have led to the use of genome sequencing and gene expression for development of multi-marker assays that show association with inflammatory state of the tumor and potential to predict response to immunotherapy. As such, melanoma serves as a model system for understanding cancer immunity and patient response to immunotherapy, either alone or in combination with other treatment modalities. Overall, the aim for the translational and clinical studies is to achieve incremental improvements through the development and identification of optimal treatment regimens, which increasingly involve doublet as well as triplet combinations, as well as through development of biomarkers to improve immune response. These and other topics in the management of melanoma were the focus of discussions at the fourth Melanoma Bridge meeting (November 29th–December 1st, 2018, Naples, Italy), which is summarised in this report.
Collapse
|
634
|
Ipenburg NA, Nieweg OE, Ahmed T, van Doorn R, Scolyer RA, Long GV, Thompson JF, Lo S. External validation of a prognostic model to predict survival of patients with sentinel node-negative melanoma. Br J Surg 2019; 106:1319-1326. [PMID: 31310333 PMCID: PMC6790583 DOI: 10.1002/bjs.11262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022]
Abstract
Background Identifying patients with sentinel node‐negative melanoma at high risk of recurrence or death is important. The European Organisation for Research and Treatment of Cancer (EORTC) recently developed a prognostic model including Breslow thickness, ulceration and site of the primary tumour. The aims of the present study were to validate this prognostic model externally and to assess whether it could be improved by adding other prognostic factors. Methods Patients with sentinel node‐negative cutaneous melanoma were included in this retrospective single‐institution study. The β values of the EORTC prognostic model were used to predict recurrence‐free survival and melanoma‐specific survival. The predictive performance was assessed by discrimination (c‐index) and calibration. Seeking to improve the performance of the model, additional variables were added to a Cox proportional hazards model. Results Some 4235 patients with sentinel node‐negative cutaneous melanoma were included. The median follow‐up time was 50 (i.q.r. 18·5–81·5) months. Recurrences and deaths from melanoma numbered 793 (18·7 per cent) and 456 (10·8 per cent) respectively. Validation of the EORTC model showed good calibration for both outcomes, and a c‐index of 0·69. The c‐index was only marginally improved to 0·71 when other significant prognostic factors (sex, age, tumour type, mitotic rate) were added. Conclusion This study validated the EORTC prognostic model for recurrence‐free and melanoma‐specific survival of patients with negative sentinel nodes. The addition of other prognostic factors only improved the model marginally. The validated EORTC model could be used for personalizing follow‐up and selecting high‐risk patients for trials of adjuvant systemic therapy.
Collapse
Affiliation(s)
- N A Ipenburg
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.,Department of Dermatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - O E Nieweg
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - T Ahmed
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| | - R van Doorn
- Department of Dermatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - R A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - G V Long
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - J F Thompson
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia.,Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - S Lo
- Melanoma Institute Australia, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
635
|
Lamichhane P, Deshmukh R, Brown JA, Jakubski S, Parajuli P, Nolan T, Raja D, Badawy M, Yoon T, Zmiyiwsky M, Lamichhane N. Novel Delivery Systems for Checkpoint Inhibitors. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E74. [PMID: 31373327 PMCID: PMC6789831 DOI: 10.3390/medicines6030074] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
Checkpoint inhibition (CPI) therapies have been proven to be powerful clinical tools in treating cancers. FDA approvals and ongoing clinical development of checkpoint inhibitors for treatment of various cancers highlight the immense potential of checkpoint inhibitors as anti-cancer therapeutics. The occurrence of immune-related adverse events, however, is a major hindrance to the efficacy and use of checkpoint inhibitors as systemic therapies in a wide range of patients. Hence, methods of sustained and tumor-targeted delivery of checkpoint inhibitors are likely to improve efficacy while also decreasing toxic side effects. In this review, we summarize the findings of the studies that evaluated methods of tumor-targeted delivery of checkpoint inhibitors, review their strengths and weaknesses, and discuss the outlook for therapeutic use of these delivery methods.
Collapse
Affiliation(s)
- Purushottam Lamichhane
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Rahul Deshmukh
- School of Pharmacy, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Julie A Brown
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Silvia Jakubski
- Department of Biostatistics, University of Florida, Gainesville, FL 32611, USA
| | - Priyanka Parajuli
- Department of Internal Medicine, Southern Illinois University, Springfield, IL 62702, USA
| | - Todd Nolan
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Dewan Raja
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Mary Badawy
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Thomas Yoon
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Mark Zmiyiwsky
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Narottam Lamichhane
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
636
|
Abstract
Anticancer immunotherapies involving the use of immune-checkpoint inhibitors or adoptive cellular transfer have emerged as new therapeutic pillars within oncology. These treatments function by overcoming or relieving tumour-induced immunosuppression, thereby enabling immune-mediated tumour clearance. While often more effective and better tolerated than traditional and targeted therapies, many patients have innate or acquired resistance to immunotherapies. Cancer immunoediting is the process whereby the immune system can both constrain and promote tumour development, which proceeds through three phases termed elimination, equilibrium and escape. Throughout these phases, tumour immunogenicity is edited, and immunosuppressive mechanisms that enable disease progression are acquired. The mechanisms of resistance to immunotherapy seem to broadly overlap with those used by cancers as they undergo immunoediting to evade detection by the immune system. In this Review, we discuss how a deeper understanding of the mechanisms underlying the cancer immunoediting process can provide insight into the development of resistance to immunotherapies and the strategies that can be used to overcome such resistance.
Collapse
|
637
|
Kennedy LB, Salama AKS. A Review of Immune-Mediated Adverse Events in Melanoma. Oncol Ther 2019; 7:101-120. [PMID: 32699983 PMCID: PMC7359990 DOI: 10.1007/s40487-019-0096-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
The use of checkpoint inhibitor-based immunotherapy has transformed the treatment landscape for melanoma as well as many other cancer types. With the ability to potentiate tumor-specific immune responses, these agents can result in durable tumor control. However, this activation of the immune system can lead to a unique constellation of side effects, distinct from other cancer therapies, collectively termed immune-mediated adverse events (irAEs). This review will focus on irAEs and guidelines for management related to the most clinically relevant checkpoint inhibitors, those that target programmed death receptor-1 (PD-1) and cytotoxic T lymphocyte antigen-4 (CTLA-4).
Collapse
|
638
|
Clinicopathologic features correlated with paradoxical outcomes in stage IIC versus IIIA melanoma patients. Melanoma Res 2019; 29:70-76. [PMID: 30169431 DOI: 10.1097/cmr.0000000000000483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Under current AJCC staging criteria, stage IIC patients paradoxically have worse outcomes than IIIA patients despite the lack of nodal metastatic disease. This study sought to identify additional clinicopathologic characteristics correlated with worse patient outcomes. Retrospective chart review of stage IIC and IIIA melanoma patients were evaluated between 1995 and 2011 with clinical follow-up through 2015. Records were reviewed for demographics, clinical characteristics, and tumor pathology. Fisher's exact test and Wilcoxon's rank-sum test were used to assess group differences. Clinicopathologic features were evaluated relative to overall survival (OS), time to distant metastases, and local/regional recurrence. Overall, 128 patients were included (45 stage IIC and 83 stage IIIA) with a median follow-up time of 5.7 years. Compared with stage IIIA patients, stage IIC patients were older, and their melanomas were more likely to be nodular, amelanotic, thicker, have higher mitotic rate, tumor lymphocytic infiltrate, no radial growth phase, and less likely to have associated precursor lesions. Stage IIC patients had shorter OS and time to distant metastases; multivariate regression revealed that older age (>55 years) and mitotic rate (>5 mitoses/mm) were independent predictors of OS. Melanomas in stage IIC disease may be biologically distinct from those that are seen in stage IIIA. While sentinel node biopsies remain the standard-of-care, these results suggest that clinicians may want to assess the clinicopathologic characteristics described above to aggressively counsel, screen for distant disease, and consider adjuvant therapy, in particular for older patients and higher mitotic rates in thicker primary tumors, regardless of nodal status.
Collapse
|
639
|
Xu H, Tan P, Zheng X, Huang Y, Lin T, Wei Q, Ai J, Yang L. Immune-related adverse events following administration of anti-cytotoxic T-lymphocyte-associated protein-4 drugs: a comprehensive systematic review and meta-analysis. Drug Des Devel Ther 2019; 13:2215-2234. [PMID: 31308633 PMCID: PMC6613615 DOI: 10.2147/dddt.s196316] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/16/2019] [Indexed: 02/05/2023] Open
Abstract
Objective: Administration of drugs targeting anti-cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is often associated with serious immune-related adverse events (irAEs). Here, we performed a comprehensive analysis of organ-specific irAEs and treatment-related hematologic abnormalities and musculoskeletal disorders resulting from anti-CTLA-4 treatment. Materials and methods: PubMed, the Cochrane library, Web of Science, and ClinicalTrials.gov were searched for studies between January 1990 and March 2018 reporting AEs associated with anti-CTLA-4 therapies. Results: A total of 11 clinical trials with 7,088 patients were included; of these, data were accessible for 10 on ClinicalTrials.gov. Compared with control therapies (placebo, chemotherapy, radiation therapy, or vaccine), anti-CTLA-4 therapies (ipilimumab and tremelimumab) were associated with an increased risk of serious irAEs, predominantly dermatologic (rash: odds ratio [OR] 3.39, P<0.01), gastrointestinal (diarrhea and colitis: OR 6.57 and 14.01, respectively; both P<0.001), endocrine (hypophysitis, hypothyroidism, adrenal insufficiency, and hypopituitarism: OR 4.22, 3.72, 3.77, and 4.73, respectively; all P<0.05), and hepatic (hepatitis, elevated alanine aminotransferase, and elevated aspartate aminotransferase: OR 4.44, 3.28, and 3.12, respectively; all P<0.05). The most common serious organ-specific irAEs were gastrointestinal (diarrhea 9.8% and colitis 5.3%). Although the incidence of selected events was higher in anti-CTLA-4-treated patients, no significant differences were found between anti-CTLA-4 and the control therapies in treatment-related hematologic abnormalities or severe musculoskeletal disorders. Conclusion: Anti-CTLA-4 therapies are associated with an increased risk of serious organ-specific irAEs, most frequently involving the gastrointestinal system; however, no increased risk of hematologic abnormalities or severe musculoskeletal disorders was detected compared with other therapies. These results underscore the need for clinical awareness and prompt and effective management of multi-organ irAEs related to anti-CTLA-4 drugs.
Collapse
Affiliation(s)
- Hang Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaonan Zheng
- West China Medical School, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yu Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
640
|
Song Y, Tieniber AD, Gimotty PA, Mitchell TC, Amaravadi RK, Schuchter LM, Fraker DL, Karakousis GC. Survival Outcomes of Patients with Clinical Stage III Melanoma in the Era of Novel Systemic Therapies. Ann Surg Oncol 2019; 26:4621-4630. [PMID: 31270717 DOI: 10.1245/s10434-019-07599-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Immune checkpoint and BRAF-targeted inhibitors have demonstrated significant survival benefits for advanced melanoma patients within the context of clinical trials. We sought to determine their impact on overall survival (OS) at a population level in order to better understand the current landscape for patients diagnosed with clinical stage III melanoma. METHODS A retrospective study was performed using the National Cancer Database. Patients diagnosed with clinical stage III melanoma were categorized by diagnosis year into two cohorts preceding the advent of novel therapies (P1: 2004-2005, P2: 2008-2009) and a contemporary group (P3: 2012-2013). OS was estimated using standard time-to-event statistical methods. RESULTS Of 3720 patients, 525 (14%) were diagnosed in P1, 1375 (37%) in P2, and 1820 (49%) in P3. Median age at diagnosis increased over time (58, 59, and 61 years in P1, P2, and P3, respectively, P = 0.004). OS increased between P2 (median 49.3 months) and P3 (median 58.2 months, Bonferroni-corrected log-rank P < 0.001) but did not differ between P1 (median 50.5 months) and P2 (Bonferroni-corrected log-rank P > 0.99). These differences persisted on multivariable analysis. OS improved for patients diagnosed in P3 compared with P1 [hazard ratio (HR) 0.76, P < 0.001] but not P2 compared with P1 (HR 0.96, P = 0.52). CONCLUSIONS OS has significantly improved nationally for patients newly diagnosed with clinical stage III melanoma in the era of novel melanoma therapies. OS outcomes will likely continue to evolve as these agents are increasingly utilized in the adjuvant setting. These data may help to better inform affected patients with respect to prognosis.
Collapse
Affiliation(s)
- Yun Song
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Andrew D Tieniber
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara C Mitchell
- Division of Hematology Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi K Amaravadi
- Division of Hematology Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Lynn M Schuchter
- Division of Hematology Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas L Fraker
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
641
|
Hu H, Dong Z, Wang X, Bai L, Lei Q, Yang J, Li L, Li Q, Liu L, Zhang Y, Ji Y, Guo L, Liu Y, Cui H. Dehydrocorydaline inhibits cell proliferation, migration and invasion via suppressing MEK1/2-ERK1/2 cascade in melanoma. Onco Targets Ther 2019; 12:5163-5175. [PMID: 31456643 PMCID: PMC6620435 DOI: 10.2147/ott.s183558] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/01/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: Alkaloids are naturally occurring chemical compounds that are widely distributed in plants, and have pharmaceutical values and low toxicity. In recent years, some of them have been demonstrated to be promising therapeutic drug candidates for cancer treatment. Herein, we tried to explore the antitumor effect of dehydrocorydaline (DHC), a natural alkaloid isolated from Corydalis, on malignant melanoma. Methods: We treated two malignant metastatic melanoma cell lines, A375 and MV3, and a normal melanocyte cell line, PIG1, with various concentrations of DHC for set amounts of time, and detected cell proliferation, migration, and invasion by using MTT, BrdU, transwell, Western blot and soft agar assay in vitro and tumorigenicity in the xenografts in vivo. Results: Our results showed that DHC dramatically blocked cell proliferation and led to cell cycle arrest at G0/G1 phase and downregulated the expressions of cell cycle regulators CDK6 and Cyclin D1 in melanoma cells. However, DHC had little inhibitory effect on normal melanocyte cell line PIG-1. Meanwhile, DHC suppressed cell invasion and migration through modulating the epithelial–mesenchymal transition (EMT) markers including E-cadherin, vimentin, as well as β-catenin. In addition, DHC also significantly attenuated tumor growth in vivo. The expressions of cell cycle-related and metastasis-related proteins were further confirmed by immunohistochemical staining in the xenografts. Importantly, MEK1/2-ERK1/2 cascade was inactivated after DHC treatment and ERK activator t-butylhydroquinone (tBHQ) treatment rescued DHC-induced cell proliferation inhibition. Conclusions: Our results indicated that DHC inhibited cell proliferation and migration/invasion via inactivating MAPK signaling, and showed that DHC might be a potential novel drug to treat malignant melanoma.
Collapse
Affiliation(s)
- Huanrong Hu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Xianxing Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Longchang Bai
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Qian Lei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Qian Li
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Lichao Liu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China
| | - Yanli Zhang
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Yacong Ji
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Leiyang Guo
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Yaling Liu
- Department of Dermatology, the Third Hospital of Hebei Medical University, Shijiazhuang 050000, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, People's Republic of China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing 400715, People's Republic of China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
642
|
Hijacking antibody-induced CTLA-4 lysosomal degradation for safer and more effective cancer immunotherapy. Cell Res 2019; 29:609-627. [PMID: 31267017 PMCID: PMC6796842 DOI: 10.1038/s41422-019-0184-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
It remains unclear why the clinically used anti-CTLA-4 antibodies, popularly called checkpoint inhibitors, have severe immunotherapy-related adverse effects (irAEs) and yet suboptimal cancer immunotherapeutic effects (CITE). Here we report that while irAE-prone Ipilimumab and TremeIgG1 rapidly direct cell surface CTLA-4 for lysosomal degradation, the non-irAE-prone antibodies we generated, HL12 or HL32, dissociate from CTLA-4 after endocytosis and allow CTLA-4 recycling to cell surface by the LRBA-dependent mechanism. Disrupting CTLA-4 recycling results in robust CTLA-4 downregulation by all anti-CTLA-4 antibodies and confers toxicity to a non-irAE-prone anti-CTLA-4 mAb. Conversely, increasing the pH sensitivity of TremeIgG1 by introducing designed tyrosine-to-histidine mutations prevents antibody-triggered lysosomal CTLA-4 downregulation and dramatically attenuates irAE. Surprisingly, by avoiding CTLA-4 downregulation and due to their increased bioavailability, pH-sensitive anti-CTLA-4 antibodies are more effective in intratumor regulatory T-cell depletion and rejection of large established tumors. Our data establish a new paradigm for cancer research that allows for abrogating irAE while increasing CITE of anti-CTLA-4 antibodies.
Collapse
|
643
|
Song Y, Bruce AN, Fraker DL, Karakousis GC. Isolated limb perfusion and infusion in the treatment of melanoma and soft tissue sarcoma in the era of modern systemic therapies. J Surg Oncol 2019; 120:540-549. [DOI: 10.1002/jso.25600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Yun Song
- Department of SurgeryHospital of the University of Pennsylvania Philadelphia Pennsylvania
| | - Adrienne N. Bruce
- Department of SurgeryHospital of the University of Pennsylvania Philadelphia Pennsylvania
| | - Douglas L. Fraker
- Department of SurgeryHospital of the University of Pennsylvania Philadelphia Pennsylvania
| | - Giorgos C. Karakousis
- Department of SurgeryHospital of the University of Pennsylvania Philadelphia Pennsylvania
| |
Collapse
|
644
|
Klemen ND, Shindorf ML, Sherry RM. Role of Surgery in Combination with Immunotherapy. Surg Oncol Clin N Am 2019; 28:481-487. [DOI: 10.1016/j.soc.2019.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
645
|
Abstract
DESIGN This was an exploratory, single-arm clinical trial that tested the immune enhancement effects of 24-weeks of Toll-like receptor 9 (TLR9) agonist (MGN1703; Lefitolimod; 60 mg × 2 weekly) therapy. METHODS We enrolled HIV-1-infected individuals on suppressive combination antiretroviral therapy. Safety was assessed throughout the study. The primary outcome was reduction in total CD4 T-cell viral DNA levels. Secondary outcomes included safety, detailed immunological and virological analyses, and time to viral rebound (viral load > 5000 copies/ml) after randomization into an analytical treatment interruption (ATI). RESULTS A total of 12 individuals completed the treatment phase and nine completed the ATI. Adverse events were limited and consistent with previous reports for MGN1703. Although the dosing regimen led to potent T-cell activation and increased HIV-1-specific T-cell responses, there were no cohort-wide changes in persistent virus (total CD4 T cells viral DNA; P = 0.34). No difference in time to rebound was observed between the ATI arms (log rank P = 0.25). One of nine ATI participants, despite harboring a large replication-competent reservoir, controlled viremia for 150 days via both HIV-1-specific cellular and antibody-mediated immune responses. CONCLUSION A period of 24 weeks of MGN1703 treatment was safe and improved innate as well as HIV-1-specific adaptive immunity in HIV-1+ individuals. These findings support the incorporation of TLR9 agonism into combination HIV-1 cure strategies. TRIAL NAME AND REGISTRATION TLR9 Enhancement of antiviral immunity in chronic HIV-1 infection: a phase 1B/2A trial; ClinicalTrials.gov NCT02443935.
Collapse
|
646
|
Assoun S, Lemiale V, Azoulay E. Molecular targeted therapy-related life-threatening toxicity in patients with malignancies. A systematic review of published cases. Intensive Care Med 2019; 45:988-997. [PMID: 31143997 PMCID: PMC7095151 DOI: 10.1007/s00134-019-05650-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Molecular targeted therapy increased overall and disease-free survival in a wide range of malignancies. Although generally well tolerated compared to chemotherapy, molecular targeted therapy may be associated with adverse events requiring ICU admission. Informing clinicians about clinical features of these toxic events might maintain awareness and favor early recognition, prompt diagnosis and treatment. METHODS We performed a systematic review of published case reports of molecular targeted therapy-related life-threatening toxicity that led to ICU admission. The search used the Pubmed database using medical subject heading (Mesh) terms, including all FDA-approved molecular targeted therapy (TT), up to March 2019. No language restriction was applied. All cases reports of patients admitted to the ICU for molecular targeted therapy-related toxicity were included. Non-FDA-approved combinations of treatments or hormonal therapy were not included. RESULTS Two hundred and fifty-three cases were identified. Nearly half of them (n = 102; 40.3%) were related to anti-angiogenic agents, mostly for gastrointestinal and cardiovascular complications. Other molecules responsible for adverse events were chiefly immune checkpoint inhibitors (n = 85, 33.6%), EGFR inhibitors (n = 33; 13.0%), and anti-HER2 (n = 10; 4.0%). They were associated with adverse events such as respiratory or hypersensitivity events. Management and outcomes associated with these life-threatening complications are reported. CONCLUSIONS Based on the vast number of treated patients, only 253 cases of molecular therapy-related severe toxicity are reported in cancer patients. Symptoms and biomarkers that depict these events need to be better identified as to allow appropriate reporting and improving dose and schedule of the treatment adapted to each patient.
Collapse
Affiliation(s)
- Sandra Assoun
- Médecine Intensive et Réanimation AP-HP, Hôpital Saint-Louis, University Paris-7 Paris-Diderot, 1 avenue Claude Vellefaux, 75010, Paris, France
| | - Virginie Lemiale
- Médecine Intensive et Réanimation AP-HP, Hôpital Saint-Louis, University Paris-7 Paris-Diderot, 1 avenue Claude Vellefaux, 75010, Paris, France
| | - Elie Azoulay
- Médecine Intensive et Réanimation AP-HP, Hôpital Saint-Louis, University Paris-7 Paris-Diderot, 1 avenue Claude Vellefaux, 75010, Paris, France.
| |
Collapse
|
647
|
Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson VG, Dalle S, Haydon A, Lichinitser M, Khattak A, Carlino MS, Sandhu S, Larkin J, Puig S, Ascierto PA, Rutkowski P, Schadendorf D, Koornstra R, Hernandez-Aya L, Di Giacomo AM, van den Eertwegh AJ, Grob JJ, Gutzmer R, Jamal R, Lorigan PC, Lupinacci R, Krepler C, Ibrahim N, Kicinski M, Marreaud S, van Akkooi AC, Suciu S, Robert C. Prognostic and predictive value of AJCC-8 staging in the phase III EORTC1325/KEYNOTE-054 trial of pembrolizumab vs placebo in resected high-risk stage III melanoma. Eur J Cancer 2019; 116:148-157. [PMID: 31200321 DOI: 10.1016/j.ejca.2019.05.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND The American Joint Committee on Cancer-8 (AJCC) classification of melanoma was implemented in January 2018. It was based on data gathered when checkpoint inhibitors were not used as adjuvant therapy in stage III melanoma. The European Organization for Research and Treatment of Cancer (EORTC) 1325/KEYNOTE-054 double-blind phase III trial evaluated pembrolizumab vs placebo in AJCC-7 stage IIIA (excluding lymph node metastasis ≤1 mm), IIIB or IIIC (without in-transit metastasis) patients after complete lymphadenectomy. PATIENTS, METHODS AND RESULTS Patients (n = 1019) were randomised 1:1 to pembrolizumab 200 mg or placebo every 3 weeks (total of 18 doses, ∼1 year). At 1.25-year median follow-up, pembrolizumab prolonged relapse-free survival (RFS) in the total population (1-year RFS rate: 75.4% vs 61.0%; hazard ratio [HR] 0.57; logrank P < 0.0001) and consistently in the AJCC-7 subgroups. Prognostic and predictive values of AJCC-8 for RFS were evaluated in this study. Patient distribution according to the AJCC-8 stage subgroups was 8% (IIIA), 34.7% (IIIB), 49.7% (IIIC), 3.7% (IIID) and 3.8% (unknown). AJCC-8 classification was strongly associated with RFS (HRs for stage IIIB, IIIC and IIID vs IIIA were 4.0, 5.7 and 12.2, respectively) but showed no predictive importance for the treatment comparison regarding RFS (test for interaction: P = 0.68). The 1-year RFS rate for pembrolizumab vs placebo and the HRs (99% confidence interval) within each AJCC-8 subgroup were as follows: stage IIIA (92.7% vs 92.5%; 0.76 [0.11-5.43]), IIIB (79.0% vs 65.5%; 0.59 [0.35-0.99]), IIIC (73.6% vs 53.9%; 0.48 [0.33-0.70]) and IIID (50.0% vs 33.3%; 0.69 [0.24-2.00]). CONCLUSIONS AJCC-8 staging had a strong prognostic importance for RFS but no predictive importance: the RFS benefit of pembrolizumab was observed across AJCC-8 subgroups in resected high-risk stage III melanoma patients.
Collapse
Affiliation(s)
| | - Christian U Blank
- Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Mario Mandala
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Mater and Royal North Shore Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Adnan Khattak
- Fiona Stanley Hospital/University of Western Australia, Perth, WA, Australia
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, Sydney, NSW, Australia
| | | | | | - Susana Puig
- Hospital Clinic Universitari de Barcelona, Barcelona, Spain
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie Institute - Oncology Center, Warsaw, Poland
| | | | - Rutger Koornstra
- Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
| | | | | | | | | | - Ralf Gutzmer
- Skin Cancer Center, Hannover Medical School, Hannover, Germany
| | - Rahima Jamal
- Centre Hospitalier de l'Université de Montréal (CHUM), Centre de recherche du CHUM, Montreal, QC, Canada
| | - Paul C Lorigan
- Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
648
|
Barquet-Munoz SA, Leitao M, Pérez Montiel MD, Santiago Concha BG. Vulvar melanoma: management of primary disease and repeated recurrences. Int J Gynecol Cancer 2019; 29:1077-1081. [PMID: 31196869 PMCID: PMC7425808 DOI: 10.1136/ijgc-2019-000610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 02/05/2023] Open
Affiliation(s)
| | - Mario Leitao
- Division of Gynecology, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York City, New York, USA
| | | | | |
Collapse
|
649
|
Hall ET, Singhal S, Dickerson J, Gabster B, Wong HN, Aslakson RA, Schapira L. Patient-Reported Outcomes for Cancer Patients Receiving Checkpoint Inhibitors: Opportunities for Palliative Care-A Systematic Review. J Pain Symptom Manage 2019; 58:137-156.e1. [PMID: 30905677 DOI: 10.1016/j.jpainsymman.2019.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/19/2022]
Abstract
CONTEXT Immune checkpoint inhibitors (ICIs) are increasingly used to treat a variety of cancers, but comparatively little is known about patient-reported outcomes (PROs) and health-related quality of life (HRQoL) among patients receiving these novel therapies. OBJECTIVES We performed a systematic review to examine PROs and HRQoL among cancer patients receiving ICIs as compared to other anticancer therapies. METHODS We systematically searched PubMed, CINAHL, Embase, Web of Science, and Scopus, using search terms representing ICIs, PROs, and HRQoL on August 10, 2018. Eligible articles were required to involve cancer patients treated with ICIs and to report PROs and/or HRQoL data. RESULTS We screened 1453 references and included 15 publications representing 15 randomized controlled trials in our analysis. Studies included several cancer types (melanoma, lung cancer, genitourinary cancer, and head/neck cancer), used four different ICIs (nivolumab, pembrolizumab, atezolizumab, and ipilimumab), and compared ICIs to a wide range of therapies (chemotherapy, targeted therapies, other immunotherapy strategies, and placebo). Studies used a total of seven different PROs to measure HRQOL, most commonly the European Organisation for the Research and Treatment of Cancer core quality of life questionnaire (EORTC QLQ-C30) (n = 12, 80%). PRO data were reported in a variety of formats and at a variety of time points throughout treatment, which made direct comparison challenging. Some trials (n = 11, 73%) reported PROs on specific symptoms. In general, patients receiving ICIs had similar-to-improved HRQoL and experiences when compared to other therapies. CONCLUSION Despite the broad clinical trials experience of ICI therapies across cancer types, relatively few randomized studies reported PROs and patient HRQoL data. Available data suggest that ICIs are well tolerated in terms of HRQoL compared to other anticancer therapies although the conclusions are limited by the heterogeneity of trial designs and outcomes. Currently used instruments may fail to capture important symptomatology unique to ICIs, underscoring a need for PROs designed specifically for ICIs.
Collapse
Affiliation(s)
- Evan T Hall
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, California, USA
| | - Surbhi Singhal
- Department of Medicine, Stanford University, Stanford, California, USA
| | - James Dickerson
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Brooke Gabster
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Hong-Nei Wong
- Lane Medical Library & Knowledge Management Center, Stanford University School of Medicine, Stanford, California, USA
| | - Rebecca A Aslakson
- Department of Medicine, Stanford University, Stanford, California, USA; Department of Anesthesiology, Stanford University, Stanford, California, USA
| | - Lidia Schapira
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, California, USA; Department of Medicine, Stanford University, Stanford, California, USA; Department of Anesthesiology, Stanford University, Stanford, California, USA.
| | | |
Collapse
|
650
|
Ruggeri RM, Campennì A, Giuffrida G, Trimboli P, Giovanella L, Trimarchi F, Cannavò S. Endocrine and metabolic adverse effects of immune checkpoint inhibitors: an overview (what endocrinologists should know). J Endocrinol Invest 2019; 42:745-756. [PMID: 30471004 DOI: 10.1007/s40618-018-0984-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 11/15/2018] [Indexed: 12/26/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are novel anticancer agents, recently introduced with the aim of boosting the immune response against tumors. ICIs are monoclonal autoantibodies that specifically target inhibitory receptors on T cells: cytotoxic T lymphocyte antigen 4 (CTLA4), programmed death 1 (PD-1) and its ligand (PD-1L). ICIs also generate peculiar dysimmune toxicities, called immune-related adverse events (irAEs), that can potentially affect any tissue, and some may be life-threatening if not promptly recognized. The endocrine and metabolic side effects of ICIs are reviewed here, with a particular focus on their clinical presentation and management. They are among the most frequent toxicities (around 10%) and include hypophysitis, thyroid disorders, adrenalitis, and diabetes mellitus. Treatment is based on the replacement of specific hormone deficits, accompanied by immunosuppression (with corticosteroids or other drugs), depending on irAEs grade, often without the need of ICI withdrawal, except in more severe forms. Prompt recognition of endocrine and metabolic irAEs and adequate treatment allow the patients to continue a therapy they are benefiting from. Endocrinologists, as an integral part of the multidisciplinary oncologic team, need to be familiar with the unique toxicity profile of these anticancer agents. Practical recommendations for their management are proposed.
Collapse
Affiliation(s)
- R M Ruggeri
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, Building H, Floor 4, "G. Martino" University Hospital, University of Messina, 98125, Messina, Italy
| | - A Campennì
- Unit of Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - G Giuffrida
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, Building H, Floor 4, "G. Martino" University Hospital, University of Messina, 98125, Messina, Italy.
| | - P Trimboli
- Nuclear Medicine, PET/CT Centre, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - L Giovanella
- Nuclear Medicine, PET/CT Centre, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - F Trimarchi
- Accademia Peloritana dei Pericolanti at the University of Messina, Messina, Italy
| | - S Cannavò
- Department of Human Pathology of Adulthood and Childhood "Gaetano Barresi", University of Messina, Messina, Italy
| |
Collapse
|