1
|
Zhou Y, Yu H, Chen C, Li A, Zhang X, Qiu H, Du W, Fu S, Zhang L, Hong S. Placebo immune-related adverse events (irAEs): A neglected phenomenon in cancer immunotherapy trials. Eur J Cancer 2024; 208:114203. [PMID: 38981314 DOI: 10.1016/j.ejca.2024.114203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE This study aims to investigate the underexplored prevalence of placebo-reported immune-related adverse events (irAEs) in immune checkpoint inhibitor (ICI) trials. METHODS We searched public databases for randomized clinical trials (RCTs) involving ICI versus placebo treatments in patients with malignancies. Study characteristics and irAEs occurrences were extracted for meta-analyses using a random-effects model. MAIN OUTCOMES Proportions of patients reported to experience any grade and grade 3 to 5 placebo irAEs; the risk ratio (RR) of reporting 'false' irAEs in the experiment arm (defined as 'false-irAE ratio', calculated by dividing the proportion of patients documented with irAEs in the placebo arm by that in the experimental arm). RESULTS 47 RCTs with 30,119 patients were analyzed. The pooled proportion of patients reported to experience any grade and grade 3 to 5 irAEs among placebo participants was 22.85 % (17.33 %-29.50 %) and 3.40 % (2.35 %-4.63 %), respectively. The pooled proportion of placebo-treated patients who experienced serious irAEs was 0.67 % (0.03 %-1.91 %). Treatment discontinuation and death due to placebo irAEs occurred in 0.69 % (<0.01 %-1.30 %) and 0.12 % (<0.01 %-0.40 %) of patients, respectively. The false-irAE ratio for any grade and grade 3 to 5 irAEs were 0.49 and 0.28. The false-irAE ratio was significantly higher in RCTs with control arms of placebo plus non-immunotherapy than in those with placebo alone (any grade: 0.57 vs. 0.32, P < 0.001; grade 3 to 5: 0.36 vs. 0.12, P = 0.009). CONCLUSION Our analyses of placebo-treated participants in ICI RCTs document the common occurrence of placebo irAEs. These findings are important for interpreting irAE profiles, avoiding inappropriate therapeutic interventions.
Collapse
Affiliation(s)
- Yixin Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP region, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Hui Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Chen Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Anlin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Huijuan Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP region, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Wei Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Sha Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou 510120, China; Department of Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou 510120, China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| |
Collapse
|
2
|
Miske R, Scharf M, Borowski K, Rieckhoff N, Teegen B, Denno Y, Probst C, Guthke K, Didrihsone I, Wildemann B, Ruprecht K, Komorowski L, Jarius S. Septin-3 autoimmunity in patients with paraneoplastic cerebellar ataxia. J Neuroinflammation 2023; 20:88. [PMID: 36997937 PMCID: PMC10061979 DOI: 10.1186/s12974-023-02718-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/03/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Septins are cytoskeletal proteins with filament forming capabilities, which have multiple roles during cell division, cellular polarization, morphogenesis, and membrane trafficking. Autoantibodies against septin-5 are associated with non-paraneoplastic cerebellar ataxia, and autoantibodies against septin-7 with encephalopathy with prominent neuropsychiatric features. Here, we report on newly identified autoantibodies against septin-3 in patients with paraneoplastic cerebellar ataxia. We also propose a strategy for anti-septin autoantibody determination. METHODS Sera from three patients producing similar immunofluorescence staining patterns on cerebellar and hippocampal sections were subjected to immunoprecipitation followed by mass spectrometry. The identified candidate antigens, all of which were septins, were expressed recombinantly in HEK293 cells either individually, as complexes, or combinations missing individual septins, for use in recombinant cell-based indirect immunofluorescence assays (RC-IIFA). Specificity for septin-3 was further confirmed by tissue IIFA neutralization experiments. Finally, tumor tissue sections were analyzed immunohistochemically for septin-3 expression. RESULTS Immunoprecipitation with rat cerebellum lysate revealed septin-3, -5, -6, -7, and -11 as candidate target antigens. Sera of all three patients reacted with recombinant cells co-expressing septin-3/5/6/7/11, while none of 149 healthy control sera was similarly reactive. In RC-IIFAs the patient sera recognized only cells expressing septin-3, individually and in complexes. Incubation of patient sera with five different septin combinations, each missing one of the five septins, confirmed the autoantibodies' specificity for septin-3. The tissue IIFA reactivity of patient serum was abolished by pre-incubation with HEK293 cell lysates overexpressing the septin-3/5/6/7/11 complex or septin-3 alone, but not with HEK293 cell lysates overexpressing septin-5 as control. All three patients had cancers (2 × melanoma, 1 × small cell lung cancer), presented with progressive cerebellar syndromes, and responded poorly to immunotherapy. Expression of septin-3 was demonstrated in resected tumor tissue available from one patient. CONCLUSIONS Septin-3 is a novel autoantibody target in patients with paraneoplastic cerebellar syndromes. Based on our findings, RC-IIFA with HEK293 cells expressing the septin-3/5/6/7/11 complex may serve as a screening tool to investigate anti-septin autoantibodies in serological samples with a characteristic staining pattern on neuronal tissue sections. Autoantibodies against individual septins can then be confirmed by RC-IIFA expressing single septins.
Collapse
Affiliation(s)
- Ramona Miske
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany
| | - Madeleine Scharf
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany.
| | - Kathrin Borowski
- Clinical Immunological Laboratory Prof. Dr. med. Winfried Stöcker, Lübeck, Germany
| | - Nicole Rieckhoff
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. Dr. med. Winfried Stöcker, Lübeck, Germany
| | - Yvonne Denno
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany
| | - Christian Probst
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany
| | - Kersten Guthke
- Department of Neurology, Städtisches Klinikum Görlitz, Görlitz, Germany
| | - Ieva Didrihsone
- Department of Neurology, Hermann-Josef-Krankenhaus, Erkelenz, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lars Komorowski
- Institute for Experimental Immunology, affiliated to EUROIMMUN AG, Lübeck, Germany
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
3
|
Rodgers CB, Mustard CJ, McLean RT, Hutchison S, Pritchard AL. A B-cell or a key player? The different roles of B-cells and antibodies in melanoma. Pigment Cell Melanoma Res 2022; 35:303-319. [PMID: 35218154 PMCID: PMC9314792 DOI: 10.1111/pcmr.13031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 02/01/2022] [Accepted: 02/21/2022] [Indexed: 12/17/2022]
Abstract
The B‐cell system plays an important role in the melanoma immune response; however, consensus has yet to be reached in many facets. Here, we comprehensively review human studies only, due to fundamental differences in the humoral response with animal models. Tumour‐infiltrating B‐cells are associated with contradictory prognostic values, reflecting a lack of agreement between studies on cell subset classification and differences in the markers used, particularly the common use of a single marker not differentiating multiple subsets. Tertiary lymphoid structures (TLS) organise T‐cells and B‐cells within tumours to generate a local anti‐tumour response and TLS presence associates with improved survival in response to immune checkpoint blockade, in late‐stage disease. Autoantibody production is increased in melanoma patients and has been proposed as biomarkers for diagnosis, prognosis and treatment/toxicity response; however, no consistent targets are yet identified. The function of antibodies in an anti‐tumour response is determined by its isotype and subclass; IgG4 is immune‐suppressive and robustly correlate with poor patient survival in melanoma. We conclude that the current B‐cell literature needs careful interpretation based on the methods used and that we need a consensus of markers to define B‐cells and associated lymphoid organs. Furthermore, future studies need to not only examine antibody targets, but also isotypes when considering functional roles.
Collapse
Affiliation(s)
- Chloe B Rodgers
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Colette J Mustard
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Ryan T McLean
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Sharon Hutchison
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Antonia L Pritchard
- Genetics and Immunology Department, Division of Biomedical Research, Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| |
Collapse
|
4
|
Eggermont A, Kicinski M, Suciu S. Association of selected (immune-related) adverse events and outcome in two adjuvant phase III trials, Checkmate-238 and EORTC1325/KEYNOTE-054. J Immunother Cancer 2022; 10:jitc-2021-004272. [PMID: 35042824 PMCID: PMC8768906 DOI: 10.1136/jitc-2021-004272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Alexander Eggermont
- Research Directorate Princess Maxima Center & Cancer Medicine University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Stefan Suciu
- Department of Statistics, EORTC, Brussels, Belgium
| |
Collapse
|
5
|
Eggermont AMM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, Dalle S, Haydon A, 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 AJM, Grob JJ, Gutzmer R, Jamal R, Lorigan PC, Krepler C, Ibrahim N, Marreaud S, van Akkooi A, Robert C, Suciu S. Association Between Immune-Related Adverse Events and Recurrence-Free Survival Among Patients With Stage III Melanoma Randomized to Receive Pembrolizumab or Placebo: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol 2020; 6:519-527. [PMID: 31895407 PMCID: PMC6990933 DOI: 10.1001/jamaoncol.2019.5570] [Citation(s) in RCA: 321] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Importance Whether immune-related adverse events (irAEs) indicate drug activity in patients treated with immune checkpoint inhibitors remains unknown. Objective To investigate the association between irAEs and recurrence-free survival (RFS) in the double-blind EORTC 1325/KEYNOTE-054 clinical trial comparing pembrolizumab therapy and placebo for the treatment of patients with high-risk stage III melanoma. Design, Setting, and Participants A total of 1019 adults with stage III melanoma were randomly assigned on a 1:1 ratio to receive treatment with pembrolizumab therapy or placebo. Eligible patients were adults 18 years and older with complete resection of cutaneous melanoma metastatic to lymph nodes, classified with stage IIIA (at least 1 micrometastasis measuring >1 mm in greatest diameter), IIIB, or IIIC (without in-transit metastasis) cancer. Patients were randomized from August 26, 2015, to November 14, 2016. The clinical cutoff for the data set was October 2, 2017. Analyses were then performed on the database, which was locked on November 28, 2017. Interventions Participants were scheduled to receive 200 mg of pembrolizumab or placebo every 3 weeks for a total of 18 doses for approximately 1 year or until disease recurrence, unacceptable toxic effects, major protocol violation, or withdrawal of consent. Main Outcomes and Measures The association between irAEs and RFS was estimated using a Cox model adjusted for sex, age, and AJCC-7 stage, with a time-varying covariate that had a value of 0 before irAE onset and 1 after irAE onset. Results Of 1011 patients who began treatment with pembrolizumab therapy or placebo, 622 (61.5%) were men and 389 (38.5%) were women; 386 patients (38.2%) were aged 50 to 64 years, 377 (37.3%) were younger than 50 years, and 248 (24.5%) were 65 years and older. Consistent with the reported main analysis in the intent-to-treat population, RFS was longer in the pembrolizumab arm compared with the placebo arm (hazard ratio [HR], 0.56; 98.4% CI, 0.43-0.74) among patients who started treatment. The incidence of irAEs was 190 (37.4%) in the pembrolizumab arm (n = 509) and 45 (9.0%) in the placebo arm (n = 502); in each treatment group, the incidence was similar for men and women. The occurrence of an irAE was associated with a longer RFS in the pembrolizumab arm (HR, 0.61; 95% CI, 0.39-0.95; P = .03) in both men and women. However, in the placebo arm, this association was not significant. Compared with the placebo arm, the reduction in the hazard of recurrence or death in the pembrolizumab arm was greater after the onset of an irAE than without or before an irAE (HR, 0.37; 95% CI, 0.24-0.57 vs HR, 0.61; 95% CI, 0.49-0.77, respectively; P = .03). Conclusions and Relevance In this study, the occurrence of an irAE was associated with a longer RFS in the pembrolizumab arm. Trial Registrations ClinicalTrials.gov identifier: NCT02362594; EudraCT identifier: 2014-004944-37.
Collapse
Affiliation(s)
| | - Michal Kicinski
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Christian U Blank
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Mario Mandala
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney and Mater and Royal North Shore Hospitals, Sydney, New South Wales, Australia
| | - Victoria Atkinson
- Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Stéphane Dalle
- Hospices Civils de Lyon Cancer Institute, Lyon University, Lyon, France
| | | | - Adnan Khattak
- Fiona Stanley Hospital, University of Western Australia, Perth, Washington, Australia
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, New South Wales, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Susana Puig
- Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie Institute Cancer Centre and Institute of Oncology, Warsaw, Poland
| | - Dirk Schadendorf
- University Hospital Essen, Essen, Germany.,Germany and German Cancer Consortium of Translational Cancer Research, Heidelberg, Germany
| | - Rutger Koornstra
- Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | | | | | | | | | - Ralf Gutzmer
- Skin Cancer Center, Hannover Medical School, Hannover, Germany
| | - Rahima Jamal
- Centre de Recherche, Centre Hospitalier de l'Universite de Montreal, Montreal, Quebec, Canada
| | - Paul C Lorigan
- Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | - Sandrine Marreaud
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Alexander van Akkooi
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Caroline Robert
- Gustave Roussy Cancer Campus Grand Paris, Universite Paris-Saclay, Villejuif, France
| | - Stefan Suciu
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| |
Collapse
|
6
|
Sorolla A, Sorolla MA, Wang E, Ceña V. Peptides, proteins and nanotechnology: a promising synergy for breast cancer targeting and treatment. Expert Opin Drug Deliv 2020; 17:1597-1613. [PMID: 32835538 DOI: 10.1080/17425247.2020.1814733] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The use of nanoparticles for breast cancer targeting and treatment has become a reality. They are safe and possess interesting peculiarities such as the unspecific accumulation into the tumor site and the possibility to activate controlled drug release as compared to free drugs. However, there are still many areas of improvement which can certainly be addressed with the use of peptide-based elements. AREAS COVERED The article reviews different preclinical strategies employing peptides and proteins in combination with nanoparticles for breast cancer targeting and treatment as well as peptide and protein-targeted encapsulated drugs, and it lists the current clinical status of therapies using peptides and proteins for breast cancer. EXPERT OPINION The conjugation of protein and peptides can improve tumor homing of nanoparticles, increase cellular penetration and attack specific drivers and vulnerabilities of the breast cancer cell to promote tumor cytotoxicity while reducing secondary effects in healthy tissues. Examples are the use of antibodies, arginylglycylaspartic acid (RGD) peptides, membrane disruptive peptides, interference peptides, and peptide vaccines. Although their implementation in the clinic has been relatively slow up to now, we anticipate great progress in the field which will translate into more efficacious and selective nanotherapies for breast cancer.
Collapse
Affiliation(s)
- Anabel Sorolla
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Maria Alba Sorolla
- Biomedical Research Institute (IRB Lleida), Research Group of Cancer Biomarkers , Lleida, Spain
| | - Edina Wang
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad De Castilla-La Mancha , Albacete, Spain.,Centro De Investigación En Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII , Madrid, Spain
| |
Collapse
|
7
|
Testori AAE, Chiellino S, van Akkooi AC. Adjuvant Therapy for Melanoma: Past, Current, and Future Developments. Cancers (Basel) 2020; 12:cancers12071994. [PMID: 32708268 PMCID: PMC7409361 DOI: 10.3390/cancers12071994] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
This review describes the progress that the concept of adjuvant therapies has undergone in the last 50 years and focuses on the most recent development where an adjuvant approach has been scientifically evaluated in melanoma clinical trials. Over the past decade the development of immunotherapies and targeted therapies has drastically changed the treatment of stage IV melanoma patients. These successes led to trials studying the same therapies in the adjuvant setting, in high risk resected stage III and IV melanoma patients. Adjuvant immune checkpoint blockade with anti-CTLA-4 antibody ipilimumab was the first drug to show an improvement in recurrence-free and overall survival but this was accompanied by high severe toxicity rates. Therefore, these results were bypassed by adjuvant treatment with anti-PD-1 agents nivolumab and pembrolizumab and BRAF-directed target therapy, which showed even better recurrence-free survival rates with more favorable toxicity rates. The whole concept of adjuvant therapy may be integrated with the new neoadjuvant approaches that are under investigation through several clinical trials. However, there is still no data available on whether the effective adjuvant therapy that patients finally have at their disposal could be offered to them while waiting for recurrence, sparing at least 50% of them a potentially long-term toxic side effect but with the same rate of overall survival (OS). Adjuvant therapy for melanoma has radically changed over the past few years—anti-PD-1 or BRAF-directed therapy is the new standard of care.
Collapse
Affiliation(s)
- Alessandro A. E. Testori
- Department of Dermatology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Correspondence: or
| | - Silvia Chiellino
- Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Alexander C.J. van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute–Antoni van Leeuwenhoek, 1066cx Amsterdam, The Netherlands;
| |
Collapse
|
8
|
Baetz TD, Fletcher GG, Knight G, McWhirter E, Rajagopal S, Song X, Petrella TM. Systemic adjuvant therapy for adult patients at high risk for recurrent melanoma: A systematic review. Cancer Treat Rev 2020; 87:102032. [PMID: 32473511 DOI: 10.1016/j.ctrv.2020.102032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022]
Abstract
Cutaneous melanoma is typically treated with wide local excision and, when appropriate, a sentinel node biopsy. Many patients are cured with this approach but for patients who have cancers with high risk features there is a significant risk of local and distant relapse and death. Interferon-based adjuvant therapy was recommended in the past but had modest results with significant toxicity. Recently, new therapies (immune checkpoint inhibitors and targeted therapies) have been found to be effective in the treatment of patients with metastatic melanoma and many of these therapies have been evaluated and found to be effective in the adjuvant treatment of high risk patients with melanoma. This systematic review of adjuvant therapies for cutaneous and mucosal melanoma was conducted for Ontario Health (Cancer Care Ontario) as the basis of a clinical practice guideline to address the question of whether patients with completely resected melanoma should be considered for adjuvant systemic therapy and which adjuvant therapy should be used.
Collapse
Affiliation(s)
- Tara D Baetz
- Department of Oncology, Queen's University, Kingston, ON, Canada; Cancer Centre of Southeastern Ontario/Kingston General Hospital, Kingston, ON, Canada.
| | - Glenn G Fletcher
- Program in Evidence-Based Care, McMaster University, Hamilton, ON, Canada
| | - Gregory Knight
- Department of Oncology, McMaster University, Hamilton, ON, Canada; Grand River Regional Cancer Centre, Kitchener, ON, Canada
| | - Elaine McWhirter
- Department of Oncology, McMaster University, Hamilton, ON, Canada; Juravinski Cancer Centre, Hamilton, ON, Canada
| | | | - Xinni Song
- Department of Internal Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada; The Ottawa Hospital Cancer Centre, Ottawa, ON, Canada
| | - Teresa M Petrella
- University of Toronto, Toronto, ON, Canada; Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
9
|
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine; Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| | - Nils-Petter Rudqvist
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
10
|
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| | - Nils-Petter Rudqvist
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
11
|
Navid F, Herzog CE, Sandoval J, Daryani VM, Stewart CF, Gattuso J, Mandrell B, Phipps S, Chemaitilly W, Sykes A, Davidoff AM, Shulkin BL, Bahrami A, Furman WL, Mao S, Wu J, Schiff D, Rao B, Pappo A. Feasibility of Pegylated Interferon in Children and Young Adults With Resected High-Risk Melanoma. Pediatr Blood Cancer 2016; 63:1207-13. [PMID: 27038395 PMCID: PMC4877209 DOI: 10.1002/pbc.25983] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/27/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Pegylated interferon α-2b (IFN α-2b) improves disease-free survival in adults with resected stage III melanoma. We conducted a study to determine the feasibility and safety of incorporating pegylated IFN α-2b as adjuvant therapy in the treatment of children and adolescents with high-risk melanoma. Pharmacokinetic studies of IFN α-2b and neuropsychological and quality of life (OL) assessments were performed. PATIENT AND METHODS Eligible patients with resected American Joint Committee on Cancer Stage IIC, IIIA, and IIIB cutaneous melanoma received nonpegylated IFN α-2b 20 million units/m(2) /day intravenously 5 days per week for 4 weeks (induction) followed by pegylated IFN α-2b 1 μg/kg/dose weekly subcutaneously (SQ) for 48 weeks (maintenance). RESULTS Twenty-three patients (15 females, median age 10 years) were enrolled. All patients completed induction therapy; five patients did not complete maintenance therapy either because of recurrent disease (n = 2) or toxicity (n = 3). The most common grade 3 and 4 toxicities of pegylated IFN α-2b were neutropenia (35%) and elevated liver transaminases (17%). The median nonpegylated IFN α-2b AUC0-∞ (5,026 pcg⋅hr/ml) was similar to adults. The median pegylated IFN α-2b exposure (48,480 pcg⋅hr/ml) was greater than the cumulative weekly exposure for nonpegylated IFN α-2b administered SQ three times per week (TIW). Validated measures demonstrated an improvement in QOL scores and no decline in psychological functioning over the course of therapy. CONCLUSIONS Pegylated IFN α-2b 1 μg/kg/dose SQ weekly as maintenance therapy in children and adolescents with high-risk melanoma is feasible with tolerable toxicity and appears to yield higher exposures than nonpegylated IFN α-2b administered SQ TIW.
Collapse
Affiliation(s)
- Fariba Navid
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN
| | | | - John Sandoval
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN,Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Vinay M. Daryani
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Clinton F. Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Jami Gattuso
- Department of Nursing Research, St. Jude Children's Research Hospital, Memphis, TN
| | - Belinda Mandrell
- Department of Nursing Research, St. Jude Children's Research Hospital, Memphis, TN
| | - Sean Phipps
- Department of Psychology, St. Jude Children's Research Hospital, Memphis, TN
| | - Wassim Chemaitilly
- Department of Pediatric Medicine-Division of Endocrinology, St. Jude Children's Research Hospital, Memphis, TN
| | - April Sykes
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Andrew M. Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN,Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Barry L. Shulkin
- Department of Radiological Sciences, St. Jude Children's Research Hospital, Memphis, TN
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Wayne L. Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN
| | - Shenghua Mao
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Jianrong Wu
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Deborah Schiff
- Department of Pediatrics, University of California-San Diego, La Jolla, CA
| | - Bhaskar Rao
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN,Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Alberto Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN,Corresponding author: Alberto Pappo, MD, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis TN 38105, Telephone: (901) 595-2322, FAX: (901) 521-9005,
| |
Collapse
|
12
|
Wong JL, Obermajer N, Odunsi K, Edwards RP, Kalinski P. Synergistic COX2 Induction by IFNγ and TNFα Self-Limits Type-1 Immunity in the Human Tumor Microenvironment. Cancer Immunol Res 2016; 4:303-11. [PMID: 26817996 DOI: 10.1158/2326-6066.cir-15-0157] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
Maintenance of CTL-, Th1-, and NK cell-mediated type-1 immunity is essential for effective antitumor responses. Unexpectedly, we observed that the critical soluble mediators of type-1 immune effector cells, IFNγ and TNFα, synergize in the induction of cyclooxygenase 2 (COX2), the key enzyme in prostaglandin (PG)E2 synthesis, and the subsequent hyperactivation of myeloid-derived suppressor cells (MDSC) within the tumor microenvironment (TME) of ovarian cancer patients. MDSC hyperactivation by type-1 immunity and the resultant overexpression of indoleamine 2,3-dioxygenase (IDO), inducible nitric oxide synthase (iNOS/NOS2), IL10, and additional COX2 result in strong feedback suppression of type-1 immune responses. This paradoxical immune suppression driven by type-1 immune cell activation was found to depend on the synergistic action of IFNγ and TNFα, and could not be reproduced by either of these factors alone. Importantly, from a therapeutic standpoint, these negative feedback limiting type-1 responses could be eliminated by COX2 blockade, allowing amplification of type-1 immunity in the ovarian cancer TME. Our data demonstrate a new mechanism underlying the self-limiting nature of type-1 immunity in the human TME, driven by the synergistic induction of COX2 by IFNγ and TNFα, and provide a rationale for targeting the COX2-PGE2 axis to enhance the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
- Jeffrey L Wong
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nataša Obermajer
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kunle Odunsi
- Departments of Gynecological Oncology and Immunology, Roswell Park Cancer Institute, Buffalo, New York
| | - Robert P Edwards
- Magee-Womens Research Institute, Ovarian Cancer Center of Excellence, Pittsburgh, Pennsylvania. Peritoneal/Ovarian Cancer Specialty Care Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania. University of Pittsburgh Cancer Institute, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania. University of Pittsburgh Cancer Institute, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
13
|
Serologic evidence of autoimmunity in E2696 and E1694 patients with high-risk melanoma treated with adjuvant interferon alfa. Melanoma Res 2014; 24:150-7. [PMID: 24509407 DOI: 10.1097/cmr.0000000000000050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We evaluated Eastern Cooperative Group phase II and III trials E2696 and E1694 to assess the incidence and prognostic significance of autoimmunity induced by adjuvant high-dose interferon-α2b (HDI). In E2696, patients with resectable high-risk melanoma were randomized to receive vaccination with GM2-KLH/QS-1 (GMK) plus concurrent HDI, GMK plus sequential HDI, or GMK alone. E1694 randomized patients to either HDI or GMK. Sera from 103 patients in E2696 and 691 patients in E1694 banked at baseline and up to three subsequent time points were tested by ELISA for the development of five autoantibodies. In E2696, autoantibodies were induced in 16 patients (23.2%; n=69) receiving HDI and GMK and two patients (5.9%; n=34) receiving GMK alone (P=0.031). Of 691 patients in E1694, 67 (19.1%) who received HDI (n=350) developed autoantibodies, but only 16 patients (4.7%) developed autoantibodies in the vaccine group (n=341; P<0.001). Almost all induced autoantibodies were detected at ≥12 weeks after the initiation of therapy. A 1-year landmark analysis among resected stage III patients treated with HDI in E1694 showed a trend toward a survival advantage associated with HDI-induced autoimmunity (hazard ratio=0.80; 95% confidence interval: 0.50-1.98; P=0.33). Therefore, adjuvant HDI therapy is associated with the induction of autoimmunity that should be further investigated prospectively as a surrogate marker of adjuvant therapeutic benefit. This potential biomarker develops over the course of up to 1 year, and cannot be used to alter the course of therapy. Studies of the genetic determinants of this response may better discriminate patients more likely to benefit from HDI immunomodulatory therapy.
Collapse
|
14
|
Di Trolio R, Simeone E, Di Lorenzo G, Buonerba C, Ascierto PA. The use of interferon in melanoma patients: a systematic review. Cytokine Growth Factor Rev 2014; 26:203-12. [PMID: 25511547 DOI: 10.1016/j.cytogfr.2014.11.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 11/11/2014] [Indexed: 11/17/2022]
Abstract
Interferon (IFN) and PEG-IFN are the only drugs approved as adjuvant therapy in patients with melanoma at high-risk of recurrence after surgical resection. Several clinical trials of adjuvant IFN, using different doses and durations of therapy, have been conducted in these patients. Results generally suggest relapse-free survival and overall survival benefits; however, questions over the optimal dose and duration of treatment and concerns over toxicity have limited its use. IFN exerts its biological activity in melanoma via multiple mechanisms of action, most of which can be considered as indirect immunomodulatory effects. As such, IFN may also be of benefit in the neoadjuvant setting, where it may have a role in melanoma patients with locally advanced disease for whom immediate surgical excision is not possible. However, this has not been well studied. The use of IFN in patients with metastatic melanoma is controversial, with limited data and no convincing evidence of a survival benefit. However, IFN therapy combined with novel biological and immunotherapies offers the potential for a synergistic effect and improved clinical outcomes. Predictive and prognostic factors to better select melanoma patients for IFN treatment have been identified (e.g. disease stage, ulceration, various cytokines) and may also enhance its therapeutic efficacy, but their incorporation into the clinical decision-making process requires validation in prospective trials. In conclusion, the modest efficacy of IFN shown in clinical trials is largely a reflection of differences in response between patients. Despite advancements in the understanding of its biological mechanisms of action, the huge potential of IFN remains to be fully explored and utilized in patients with melanoma.
Collapse
Affiliation(s)
- Rossella Di Trolio
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy.
| | - Ester Simeone
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy.
| | - Giuseppe Di Lorenzo
- Oncology Division, Department of Clinical Medicine, University "Federico II" of Naples, Italy.
| | - Carlo Buonerba
- Oncology Division, Department of Clinical Medicine, University "Federico II" of Naples, Italy.
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy.
| |
Collapse
|
15
|
Emerging clinical issues in melanoma in the molecularly targeted era. Methods Mol Biol 2014; 1102:11-26. [PMID: 24258971 DOI: 10.1007/978-1-62703-727-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The standard of care of patients with malignant melanoma is dramatically changing, hallmarked by the approval of three new agents for the treatment of malignant melanoma in 2011. In this changing therapeutic landscape, several clinical issues are emerging which will best be addressed through the application of advances in molecular analytics, diagnostics, and therapeutics. It is expected that dedicated and coordinated efforts in basic, translational, and clinical will be responsible for the next major breakthroughs in the care of patients with this dreaded disease. In this chapter, five critical, emerging clinical issues are presented with descriptions of approaches that might be expected to help solve these challenges to optimal patient care.
Collapse
|
16
|
Zörnig I, Halama N, Lorenzo Bermejo J, Ziegelmeier C, Dickes E, Migdoll A, Kaiser I, Waterboer T, Pawlita M, Grabe N, Ugurel S, Schadendorf D, Falk C, Eichmüller SB, Jäger D. Prognostic significance of spontaneous antibody responses against tumor-associated antigens in malignant melanoma patients. Int J Cancer 2014; 136:138-51. [PMID: 24839182 DOI: 10.1002/ijc.28980] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 04/24/2014] [Indexed: 11/06/2022]
Abstract
Distribution, patterns and prognostic impact of spontaneous antibody responses against different tumor-associated antigens (TAAs) in malignant melanoma patients are unknown so far and were investigated in this study for the first time in a large cohort at different stages of the disease, identifying new prognostic biomarkers for malignant melanoma. Serum samples from 365 melanoma patients (97 Stage I melanoma patients, 87 Stage II, 92 Stage III and 89 Stage IV) and 100 age and gender matched healthy control donors were analyzed. Samples were drawn at the time of diagnosis (Stages I-III) or at time of diagnosis of distant metastasis (Stage IV). Applying a novel multiplex assay, humoral immune responses against 29 TAAs were determined and the association between response and patient survival was investigated. Antibody responses were mainly found in melanoma patients and all tested antigens elicited immune responses in all disease stages. Antibody responses against single antigens were either associated with poor prognosis and/or shorter progression-free survival (PFS) or had no influence. While in Stages I-III significant associations were observed between an antibody response and overall survival or PFS, among Stage IV patients, no significant association was found. Multivariate analyses identified specific humoral immune responses as prognostic factors independently of age, chemotherapy and immunotherapy. Antibody responses against specific TAA in Stage I-III melanoma patients correlate with poor prognosis and/or shorter PFS. These results may help to design clinical studies in order to evaluate the potential of these responses as prognostic serological biomarkers.
Collapse
Affiliation(s)
- Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
In the past decade, major advances have been made in the understanding of melanoma. New predisposition genes have been reported and key somatic events, such as BRAF mutation, directly translated into therapeutic management. Surgery for localised melanoma and regional lymph node metastases is the standard of care. Sentinel-node biopsy provides precise staging, but has not been reported to affect survival. The effect of lymph-node dissection on survival is a topic of investigation. Two distinct approaches have emerged to try to extend survival in patients with metastatic melanoma: immunomodulation with anti-CTLA4 monoclonal antibodies, and targeted therapy with BRAF inhibitors or MEK inhibitors for BRAF-mutated melanoma. The combination of BRAF inhibitors and MEK inhibitors might improve progression-free survival further and, possibly, increase overall survival. Response patterns differ substantially-anti-CTLA4 immunotherapy can induce long-term responses, but only in a few patients, whereas targeted drugs induce responses in most patients, but nearly all of them relapse because of pre-existing or acquired resistance. Thus, the long-term prognosis of metastatic melanoma remains poor. Anti-PD1 and anti-PDL1 antibodies have emerged as breakthrough drugs for melanoma that have high response rates and long durability. Biomarkers that have predictive value remain elusive in melanoma, although emerging data for adjuvant therapy indicate that interferon sensitivity is associated with ulceration of the primary melanoma. Intense investigation continues for clinical and biological markers that predict clinical benefit of immunotherapeutic drugs, such as interferon alfa or anti-CTLA4 antibodies, and the mechanisms that lead to resistance of targeted drugs.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France; Erasmus University Medical Centre, Rotterdam, Netherlands.
| | - Alan Spatz
- Department of Pathology, McGill University & Lady Davis Institute for Medical Research, Montreal, QC, Canada
| | - Caroline Robert
- Melanoma Unit and INSERM U981, Gustave Roussy Cancer Institute, Grand Paris, Villejuif, France
| |
Collapse
|
18
|
Davar D, Tarhini AA, Gogas H, Kirkwood JM. Advances in adjuvant therapy: potential for prognostic and predictive biomarkers. Methods Mol Biol 2014; 1102:45-69. [PMID: 24258973 PMCID: PMC8591980 DOI: 10.1007/978-1-62703-727-3_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Melanoma is the third most common skin cancer but accounts for the majority of skin cancer-related mortality. The rapidly rising incidence and younger age at diagnosis has made melanoma a leading cause of lost productive years of life and has increased the urgency of finding improved adjuvant therapy for melanoma. Interferon-α was approved for the adjuvant treatment of resected high-risk melanoma following studies that demonstrated improvements in relapse-free survival and overall survival that were commenced nearly 30 years ago. The clinical benefits associated with this agent have been consistently observed across multiple studies and meta-analyses in terms of relapse rate, and to a smaller and less-consistent degree, mortality. However, significant toxicity and lack of prognostic and/or predictive biomarkers that would allow greater risk-benefit ratio have limited the more widespread adoption of this modality.Recent success with targeted agents directed against components of the MAP-kinase pathway and checkpoint inhibitors have transformed the treatment landscape in metastatic disease. Current research efforts are centered around discovering predictive/prognostic biomarkers and exploring the options for more effective regimens, either singly or in combination.
Collapse
Affiliation(s)
- Diwakar Davar
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
19
|
Torino F, Barnabei A, Paragliola R, Baldelli R, Appetecchia M, Corsello SM. Thyroid dysfunction as an unintended side effect of anticancer drugs. Thyroid 2013; 23:1345-66. [PMID: 23750887 DOI: 10.1089/thy.2013.0241] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several of the currently used anticancer drugs may variably affect thyroid function, with impairment ranging from modified total but not free concentration of thyroid hormones to overt thyroid disease. SUMMARY Cytotoxic agents seem to alter thyroid function in a relatively small proportion of adult patients. Anticancer hormone drugs may mainly alter serum levels of thyroid hormone-binding proteins without clinically relevant thyroid dysfunction. Old immunomodulating drugs, such as interferon-α and interleukin-2, are known to induce variably high incidence of autoimmune thyroid dysfunction. Newer immune checkpoint inhibitors, such as anti-CTLA4 monoclonal antibodies, are responsible for a relatively low incidence of thyroiditis and may induce secondary hypothyroidism resulting from hypophysitis. Central hypothyroidism is a well-recognized side effect of bexarotene. Despite their inherent selectivity, tyrosine kinase inhibitors may cause high rates of thyroid dysfunction. Notably, thyroid toxicity seems to be restricted to tyrosine kinase inhibitors targeting key kinase-receptors in angiogenic pathways, but not other kinase-receptors (e.g., epidermal growth factor receptors family or c-KIT). In addition, a number of these agents may also increase the levothyroxine requirement in thyroidectomized patients. CONCLUSIONS The pathophysiology of thyroid toxicity induced by many anticancer agents is not fully clarified and for others it remains speculative. Thyroid dysfunction induced by anticancer agents is generally manageable and dose reduction or discontinuation of these agents is not required. The prognostic relevance of thyroid autoimmunity, overt and subclinical hypothyroidism induced by anticancer drugs, the value of thyroid hormone replacement in individuals with abnormal thyrotropin following anticancer systemic therapy, and the correct timing of replacement therapy in cancer patients need to be defined more accurately in well-powered prospective clinical trials.
Collapse
Affiliation(s)
- Francesco Torino
- 1 Department of Systems Medicine, Tor Vergata University of Rome , Rome, Italy
| | | | | | | | | | | |
Collapse
|
20
|
Eggermont AMM. Adjuvant therapy for high-risk melanoma. EJC Suppl 2013. [PMID: 26217119 PMCID: PMC4041312 DOI: 10.1016/j.ejcsup.2013.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
21
|
Update on the role of ipilimumab in melanoma and first data on new combination therapies. Curr Opin Oncol 2013; 25:166-72. [PMID: 23299197 DOI: 10.1097/cco.0b013e32835dae4f] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW This article provides an update on the therapeutic role of the monoclonal antibody ipilimumab in melanoma. Recent therapeutic combinations, as well as directions for further investigations, will also be discussed. RECENT FINDINGS By blocking the interaction between CTLA-4 and B7 expressed on activated T lymphocytes and antigen-presenting cells, respectively, ipilimumab inhibits negative signals that physiologically downregulate T-cell activation and exerts its therapeutic activity by upregulating the antitumor activity of T lymphocytes. Ipilimumab has been the first agent to significantly improve the survival of metastatic melanoma patients and to provide long-term benefit to a sizeable proportion of patients treated within phase II/III studies and expanded access programs. On these premises, a number of studies combining ipilimumab with cytotoxic, antiangiogenic, and targeted agents have been most recently conducted. SUMMARY Ipilimumab is the prototype of a growing family of 'immunomodulating antibodies' and it has demonstrated that immunotherapy will play an increasingly important role in the new treatment approaches for cancer. Combinations of chemotherapy, radiation therapy, and targeted drugs with ipilimumab indicate that additive and synergistic antitumor activity can be achieved. Most importantly, they indicate that involving the immune system is a key strategy to improve the outcome in cancer patients.
Collapse
|
22
|
Abstract
The potential for guarantee-time bias (GTB), also known as immortal time bias, exists whenever an analysis that is timed from enrollment or random assignment, such as disease-free or overall survival, is compared across groups defined by a classifying event occurring sometime during follow-up. The types of events associated with GTB are varied and may include the occurrence of objective disease response, onset of toxicity, or seroconversion. However, comparative analyses using these types of events as predictors are different from analyses using baseline characteristics that are specified completely before the occurrence of any outcome event. Recognizing the potential for GTB is not always straightforward, and it can be challenging to know when GTB is influencing the results of an analysis. This article defines GTB, provides examples of GTB from several published articles, and discusses three analytic techniques that can be used to remove the bias: conditional landmark analysis, extended Cox model, and inverse probability weighting. The strengths and limitations of each technique are presented. As an example, we explore the effect of bisphosphonate use on disease-free survival (DFS) using data from the BIG (Breast International Group) 1-98 randomized clinical trial. An analysis using a naive approach showed substantial benefit for patients who received bisphosphonate therapy. In contrast, analyses using the three methods known to remove GTB showed no statistical evidence of a reduction in risk of a DFS event with bisphosphonate therapy.
Collapse
|
23
|
Busse A, Rapion J, Fusi A, Suciu S, Nonnenmacher A, Santinami M, Kruit WHJ, Testori A, Punt CJA, Dalgleish AG, Spatz A, Eggermont AMM, Keilholz U. Analysis of surrogate gene expression markers in peripheral blood of melanoma patients to predict treatment outcome of adjuvant pegylated interferon alpha 2b (EORTC 18991 side study). Cancer Immunol Immunother 2013; 62:1223-33. [PMID: 23624802 PMCID: PMC11028414 DOI: 10.1007/s00262-013-1428-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 04/10/2013] [Indexed: 10/26/2022]
Abstract
We analysed mRNA levels of interferon response genes (ISG15, STAT1, CXCL10) of inhibitors of the JAK/STAT pathway (STAT3, SOCS1, SOCS3) and of cytokines (TNFα, IL10, TGFß1) in peripheral blood of 91 stage III melanoma patients enrolled in EORTC 18991 trial to find biomarkers indicative for disease stage and predictive for efficacy of pegylated interferon alpha-2b (PEG-IFNα-2b) therapy. mRNA levels were analysed at baseline and after 6 months. Univariate and multivariate analyses were performed to estimate the prognostic and predictive role of mRNA levels for distant metastasis-free survival (DMFS) and relapse-free survival (RFS). Compared to healthy controls, melanoma patients showed significantly higher TGFβ1 mRNA levels. In a multivariate model, increasing SOCS1 and SOCS3 mRNA levels were associated with worse RFS (P = 0.02 and P = 0.04, respectively) and DMFS (P = 0.05 and P = 0.05, respectively) due to negative correlation between, respectively, SOCS1/SOCS3 mRNA levels and ulceration or Breslow thickness. No impact of PEG-IFNα-2b on mRNA levels was observed except for ISG15 mRNA levels, which decreased in the treatment arm (P = 0.001). It seems that patients with a decrease >60 % of ISG15 mRNA levels during 6 months PEG-IFNα-2b had inferior outcome.
Collapse
Affiliation(s)
- Antonia Busse
- Department of Medicine III, Charité-CBF, Hindenburgdamm 30, 12200, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kirkwood JM, Davar D, Tarhini A. Adjuvant immunotherapy of melanoma and development of new approaches using the neoadjuvant approach. Clin Dermatol 2013; 31:237-50. [PMID: 23608443 PMCID: PMC3654101 DOI: 10.1016/j.clindermatol.2012.08.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanoma is the third most common skin cancer but the leading cause of death from cutaneous malignancies. Although early-stage disease is frequently cured by surgical resection with excellent long-term survival, patients with deeper primary lesions (AJCC stage IIB-C) and those with microscopic (IIIA) or clinically evident regional lymph node or in-transit metastases (IIIB-C) have an increased risk of relapse and death, the latter approaching 70% or more at 5 years. In patients at high risk of recurrence/metastases, adjuvant therapy with high-dose interferon alpha-2b (HDI) following definitive surgical resection has been shown to improve relapse-free and overall survival. Neoadjuvant chemotherapy and/or radiotherapy have offered the prospect to improve regional recurrence risk and overall survival in several solid tumors. The advent of effective new molecularly targeted therapies for metastatic disease and new immunotherapies that overcome checkpoints of immune response have augmented the range of new options that are in current trial evaluation to determine their role as potential adjuvant therapies, alone and in combination with one another, and the established modality of IFN-α. The differential characteristics of the host immune response between early and advanced melanoma provide a strong mechanistic rationale for the use of neoadjuvant immunotherapeutic approaches in melanoma, and the opportunity to evaluate the mechanism of action suggest neoadjuvant trial evaluation for each of the new candidate agents and combinations of interest. Several neoadjuvant trials have been conducted in the phase II setting, which have illuminated the mechanism of IFN-α, as well as providing insight to the effects of anti-CTLA4 blocking antibodies. These agents (anti-CTLA4 blocking antibody ipilimumab, and BRAF inhibitor vemurafenib) are likely to be followed by other immunotherapies that may overcome the PD-1 checkpoint (anti-PD1 and anti-PDL-1) as well as other molecularly targeted agents such as the BRAF inhibitor dabrafenib and the MEK inhibitors trametinib, selumetinib, and MEK162 in the near future. Evaluation of the clinical role of these agents as adjuvant therapy will take years to accomplish to ascertain the relapse-free survival benefits and overall survival benefits of these agents, but neoadjuvant exploration may provide early critical evidence of the therapeutic benefits, as well as clarifying the mechanisms of these agents alone and in combination.
Collapse
Affiliation(s)
- John M. Kirkwood
- Professor of Medicine, Dermatology and Translational Science, Division of Hematology-Oncology, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA 15232, , Phone: 412-623-7707, Fax: 412-623-7704
| | - Diwakar Davar
- Division of General Internal Medicine, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, , Pager: 412-263-7622
| | - Ahmad Tarhini
- Assistant Professor of Medicine, Clinical and Translational Science, Division of Hematology-Oncology, University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh PA 15232,
| |
Collapse
|
25
|
Körner R, Preuss KD, Fadle N, Madjidi D, Neumann F, Bergeler L, Gräber S, Müller CSL, Grünhage F, Pfreundschuh M, Lammert F, Vogt T, Pföhler C. Serum antibodies against CD28-- a new potential marker of dismal prognosis in melanoma patients. PLoS One 2013; 8:e58087. [PMID: 23483974 PMCID: PMC3590176 DOI: 10.1371/journal.pone.0058087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 01/30/2013] [Indexed: 12/02/2022] Open
Abstract
Background Autoantibodies against CD28 have been found in patients with autoimmune and atopic diseases. These antibodies may act as superagonists and activate T cells but may also be antagonistic or induce immunosuppressive effects by activating regulatory T cells. Autoimmunity in melanoma patients has been discussed controversially. Objective We investigated 230 melanoma patients for the occurrence of CD28 antibodies and the effect of the latter on overall and progress-free survival. Methods We constructed an ELISA assay to measure CD28 serum antibodies. 230 patients with melanoma and a control-group of 625 patients consistent of 212 patients with virus hepatitis b or c, 149 patients with allergies, 78 patients with psoriasis, 46 patients with plasmocytoma and 140 healthy blood donors were investigated for the occurrence of CD28 antibodies. Results CD28 abs occur at a higher percentage in patients with melanoma and in patients with viral hepatitis than in other groups investigated (p<0.001). Occurrence of CD28 abs is significantly higher in patients receiving interferons independent from the underlying disease (p<0.001). In vitro CD28 serum antibodies have an inhibitory effect on the CD28 receptor as they lead to reduced stimulation of Jurkat cells. Presence of CD28 was correlated with a higher risk of dying from melanoma (p = 0.043), but not with a significantly shortened overall survival or progression-free survival. Conclusion Interferon therapy appears to induce the production of CD28 abs. In light of reports that these CD28 abs induce immunosuppressive Tregs and – as our data show – that they are inhibitors of CD28 receptor mediated stimulation, the continuation of therapies with interferons in melanoma patients developing CD28 antibodies should be critically reconsidered, since our data indicate a worse outcome of patients with CD28 abs.
Collapse
Affiliation(s)
- Rebecca Körner
- Department of Dermatology, Saarland University Hospital, Homburg/Saar, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Becquart C, Ryckewaert G, Desmedt E, Defebvre L, Le Rhun E, Mortier L. [Limbic encephalitis: a new paraneoplastic auto-immune manifestation associated with metastatic melanoma?]. Ann Dermatol Venereol 2013; 140:278-81. [PMID: 23567229 DOI: 10.1016/j.annder.2013.01.424] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 12/01/2012] [Accepted: 01/11/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND Several studies indicate an association between immune-related manifestations and prolonged survival in metastatic melanoma. Limbic encephalitis driven by immune-mediated disorders may also be observed during the course of certain cancers. PATIENTS AND METHODS In November 2009, a 60-year-old woman followed up for metastatic melanoma since July 2005 developed rapidly progressive cognitive disorder. Clinical, biological, MRI and electroencephalogram abnormalities resulted in diagnosis of probably paraneoplastic limbic encephalitis in a context of immune-related manifestations although chemotoxicity could not be ruled out. Auto-immunity with hypothyroidism and thrombocytopenia were seen concomitantly. DISCUSSION To the best of our knowledge, this is the first reported case of probably paraneoplastic limbic encephalitis associated with melanoma, a new example of an immune-related condition associated with prolonged survival in metastatic melanoma.
Collapse
Affiliation(s)
- C Becquart
- Service de dermatologie, hôpital Claude-Huriez, rue Michel-Polonovski, 59037 Lille cedex, France.
| | | | | | | | | | | |
Collapse
|
27
|
Moreno Nogueira JA, Valero Arbizu M, Pérez Temprano R. Adjuvant treatment of melanoma. ISRN DERMATOLOGY 2013; 2013:545631. [PMID: 23476798 PMCID: PMC3588212 DOI: 10.1155/2013/545631] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 12/02/2012] [Indexed: 11/17/2022]
Abstract
Melanomas represent 4% of all malignant tumors of the skin, yet account for 80% of deaths from skin cancer.While in the early stages patients can be successfully treated with surgical resection, metastatic melanoma prognosis is dismal. Several oncogenes have been identified in melanoma as BRAF, NRAS, c-Kit, and GNA11 GNAQ, each capable of activating MAPK pathway that increases cell proliferation and promotes angiogenesis, although NRAS and c-Kit also activate PI3 kinase pathway, including being more commonly BRAF activated oncogene. The treatment of choice for localised primary cutaneous melanoma is surgery plus lymphadenectomy if regional lymph nodes are involved. The justification for treatment in addition to surgery is based on the poor prognosis for high risk melanomas with a relapse index of 50-80%. Patients included in the high risk group should be assessed for adjuvant treatment with high doses of Interferon- α 2b, as it is the only treatment shown to significantly improve disease free and possibly global survival. In the future we will have to analyze all these therapeutic possibilities on specific targets, probably associated with chemotherapy and/or interferon in the adjuvant treatment, if we want to change the natural history of melanomas.
Collapse
Affiliation(s)
- J. A. Moreno Nogueira
- Department of Oncology, Virgen del Rocio University Hospital, Royal Academy of Medicine, 41001 Seville, Spain
| | - M. Valero Arbizu
- Department of Oncology, Infanta Luisa Hospital, 41010 Seville, Spain
| | - R. Pérez Temprano
- Department of Medicine, Virgen Macarena University Hospital, 41014 Seville, Spain
| |
Collapse
|
28
|
Ascierto PA, Gogas HJ, Grob JJ, Algarra SM, Mohr P, Hansson J, Hauschild A. Adjuvant interferon alfa in malignant melanoma: An interdisciplinary and multinational expert review. Crit Rev Oncol Hematol 2013; 85:149-61. [DOI: 10.1016/j.critrevonc.2012.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/28/2012] [Accepted: 07/10/2012] [Indexed: 11/25/2022] Open
|
29
|
Jarius S, Steinmeyer F, Knobel A, Streitberger K, Hotter B, Horn S, Heuer H, Schreiber SJ, Wilhelm T, Trefzer U, Wildemann B, Ruprecht K. GABAB receptor antibodies in paraneoplastic cerebellar ataxia. J Neuroimmunol 2013; 256:94-6. [PMID: 23332614 DOI: 10.1016/j.jneuroim.2012.12.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/08/2012] [Accepted: 12/12/2012] [Indexed: 11/29/2022]
Abstract
Autoantibodies to the gamma-aminobutyric acid-B (GABAB) receptor were recently described in patients with limbic encephalitis presenting with early or prominent seizures. We report on a 64-year-old man with malignant melanoma who during adjuvant therapy with interferon (IFN)-alpha developed cerebellar ataxia. Indirect immunofluorescence on brain tissue sections revealed high-titer (1:20,000) IgG1 serum autoantibodies to the cerebellar molecular and granular layer, which were confirmed to be directed against GABAB receptor in a cell-based assay. This case highlights cerebellar ataxia in the absence of seizures as a clinical manifestation of GABAB receptor autoimmunity and extends the spectrum of tumors underlying this condition to malignant melanoma. IFN-alpha therapy may have contributed to the development of autoimmunity in this patient.
Collapse
Affiliation(s)
- Sven Jarius
- Division of Molecular Neuroimmunology, Department of Neurology, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Eggermont AM, Suciu S, Testori A, Santinami M, Kruit WH, Marsden J, Punt CJ, Salès F, Dummer R, Robert C, Schadendorf D, Patel PM, de Schaetzen G, Spatz A, Keilholz U. Long-Term Results of the Randomized Phase III Trial EORTC 18991 of Adjuvant Therapy With Pegylated Interferon Alfa-2b Versus Observation in Resected Stage III Melanoma. J Clin Oncol 2012; 30:3810-8. [DOI: 10.1200/jco.2011.41.3799] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Adjuvant pegylated interferon alfa-2b (PEG-IFN-α-2b) was approved for treatment of resected stage III melanoma in 2011. Here, we present long-term follow-up results of this pivotal trial. Patients and Methods In all, 1,256 patients with resected stage III melanoma were randomly assigned to observation (n = 629) or PEG-IFN-α-2b (n = 627) for an intended duration of 5 years. Stratification factors were microscopic (N1) versus macroscopic (N2) nodal involvement, number of positive nodes, ulceration and tumor thickness, sex, and center. Recurrence-free survival (RFS; primary end point), distant metastasis-free survival (DMFS), and overall survival (OS) were analyzed for the intent-to-treat population. Results At 7.6 years median follow-up, 384 recurrences or deaths had occurred with PEG-IFN-α-2b versus 406 in the observation group (hazard ratio [HR], 0.87; 95% CI, 0.76 to 1.00; P = .055); 7-year RFS rate was 39.1% versus 34.6%. There was no difference in OS (P = .57). In stage III-N1 ulcerated melanoma, RFS (HR, 0.72; 99% CI, 0.46 to 1.13; P = .06), DMFS (HR, 0.65; 99% CI, 0.41 to 1.04; P = .02), and OS (HR, 0.59; 99% CI, 0.35 to 0.97; P = .006) were prolonged with PEG-IFN-α-2b. PEG-IFN-α-2b was discontinued for toxicity in 37% of patients. Conclusion Adjuvant PEG-IFN-α-2b for stage III melanoma had a positive impact on RFS, which was marginally significant and slightly diminished versus the benefit seen at prior follow-up (median, 3.8 years). No significant increase in DMFS or OS was noted in the overall population. Patients with ulcerated melanoma and lower disease burden had the greatest benefit.
Collapse
Affiliation(s)
- Alexander M.M. Eggermont
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Stefan Suciu
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Alessandro Testori
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Mario Santinami
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Wim H.J. Kruit
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Jeremy Marsden
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Cornelis J.A. Punt
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - François Salès
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Reinhard Dummer
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Caroline Robert
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Dirk Schadendorf
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Poulam M. Patel
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Gaetan de Schaetzen
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Alan Spatz
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| | - Ulrich Keilholz
- Alexander M.M. Eggermont and Caroline Robert, Institut de Cancérologie Gustave Roussy, Villejuif, France; Stefan Suciu and Gaetan de Schaetzen, European Organisation for Research and Treatment of Cancer Headquarters; François Salès, Institut Jules Bordet, Brussels, Belgium; Alessandro Testori, Istituto Europeo di Oncologia; Mario Santinami, Istituto Nazionale dei Tumori, Milan, Italy; Wim H.J. Kruit, Erasmus University Medical Center, Rotterdam; Cornelis J.A. Punt, Academic Medical Center, Amsterdam, the
| |
Collapse
|
31
|
Abstract
Even early clinical studies showed that adjuvant chemotherapy achieved no therapeutic benefit for melanomas so that in the current guidelines its use is only recommended within the framework of clinical studies. For over 30 years interferons have been used in the adjuvant treatment of primary high risk melanomas as well as in the treatment of metastasized melanomas. They function in an antiviral, immune modulating and antitumor fashion. Direct and indirect effects on tumor cells could be demonstrated for interferons. In Europe low dosage interferon therapy is approved and has become widely established for stage II melanomas, whereas in the USA high dosage therapy for stage III and since March 2011 therapy with pegylated interferon in stage III are also approved. In this article the most important study results will be dealt with in detail. In summary, according to the current study situation therapy with interferon should be offered especially to patients with ulcerated primary melanoma and microscopic lymph node infiltration. Many attempts have been made in the last decades to positively influence the survival time of distant metastasized melanoma by systemic therapy. The recent development of the antibody ipilimumab against cytotoxic T-lymphocyte protein 4 (CTLA-4) could show for the first time a survival advantage in the therapy of melanoma patients in advance stage disease. The licensing of ipilimumab has meant that there is now a new standard available for the second line therapy of malignant melanoma which will be included in the guidelines on therapy of malignant melanoma. A further interesting option for adjuvant therapy is currently vaccination with the recombinant melanoma-associated protein 3 (MAGE-A3) protein in combination with the adjuvant AS015.
Collapse
|
32
|
Abstract
Estimates from the U.S. Surveillance, Epidemiology, and End Results registry suggest that melanoma incidence will reach 70,230 cases in 2011, of whom 8790 will die. The rising incidence and predilection for young individuals makes this tumor a leading source of lost productive years in the society.High-dose interferon-α-2b is the only agent approved for adjuvant therapy for melanoma; the improvement in relapse-free survival has been observed across nearly all published studies and meta-analyses. However, toxicity affects compliance, and current research is focusing on biomarkers that may allow selection of patients with greater likelihood of response and exploring new agents either singly or in combination that may improve on the benefit of interferon.In this article, we review the data for the adjuvant therapy for malignant melanoma--focusing on the results obtained with various regimens testing the several formulations of interferon-α2 and the adjuvant studies of vaccines and radiotherapy. Recent advances in the treatment of metastatic disease have established a role for CTLA-4 blockade and BRAF-inhibition, raising hopes that these agents may have a role in the adjuvant setting. At present, several trials investigating combinations of novel agents with existing immunomodulators are underway.
Collapse
|
33
|
Márquez-Rodas I, Martín Algarra S, Avilés Izquierdo JA, Custodio Cabello S, Martín M. A new era in the treatment of melanoma: from biology to clinical practice. Clin Transl Oncol 2012; 13:787-92. [PMID: 22082642 DOI: 10.1007/s12094-011-0734-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Melanoma is the deadliest cutaneous malignancy and its incidence continues to grow. Until 2011, the treatment options for metastatic melanoma were scarce and without any overall survival benefit. The emergence of new targeted therapies for BRAF mutant melanoma (vemurafenib) and immunotherapy (ipilimumab) has changed the standard of care for this disease. The objective of the present review is to summarise the biological background of the new therapeutic approaches in melanoma, focusing on apoptosis resistance, immune modulation and angiogenesis, and the direct translation into clinical practice.
Collapse
Affiliation(s)
- I Márquez-Rodas
- Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañón, Universidad Complutense, Madrid, Spain.
| | | | | | | | | |
Collapse
|
34
|
Patel JN, Walko CM. Sylatron: a pegylated interferon for use in melanoma. Ann Pharmacother 2012; 46:830-8. [PMID: 22619474 DOI: 10.1345/aph.1q791] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To review the currently available literature on peginterferon alfa-2b (pegIFN [Sylatron]), including its role in therapy and toxicity for adjuvant treatment of locally advanced melanoma. DATA SOURCES A literature search was performed of PubMed and the American Society of Clinical Oncology abstracts from 1976 to February 2012, using the primary search terms peginterferon alfa-2b, interferon, Sylatron, and melanoma. STUDY SELECTION AND DATA EXTRACTION All available English-language articles and trials that described the pharmacology, pharmacokinetics, pharmacodynamics, clinical activity, or safety profile of pegIFN were reviewed. DATA SYNTHESIS PegIFN was approved in March 2011 for the adjuvant treatment of node-positive melanoma. Interferon (IFN) is commonly used in patients with melanoma who remain at high risk for relapse following surgery; however, the optimal scheduling and dose are not agreed upon. Pegylation of IFN involves conjugation with polyethylene glycol. Following subcutaneous injection of pegIFN, the rate of absorption, renal and cellular clearance, and immunogenicity are reduced. As a result of the extended serum half-life, once-weekly administration is feasible, compared with the daily and/or thrice weekly dosing of IFN. When compared with observation alone in patients with resected stage III melanoma, pegIFN demonstrated a significant increase in relapse-free survival, with a marginal impact on overall survival. The most common adverse events were as expected with IFN and included fatigue, increased liver enzymes, pyrexia, headache, anorexia, myalgia, nausea, chills, depression, and injection site reactions. A large Phase 3 study is underway to further assess outcome and toxicity differences between pegIFN weekly and low-dose IFN thrice weekly. CONCLUSIONS PegIFN is a modified version of the previously approved interferon indicated for the adjuvant treatment of melanoma. Although the safety profile remains similar between the pegylated and non-pegylated forms, once-weekly administration is feasible secondary to an extended serum half-life and may have improved convenience for the patient.
Collapse
Affiliation(s)
- Jai N Patel
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
35
|
Is ulceration in cutaneous melanoma just a prognostic and predictive factor or is ulcerated melanoma a distinct biologic entity? Curr Opin Oncol 2012; 24:137-40. [DOI: 10.1097/cco.0b013e32834fcb0d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Testori A, Suciu S, van Akkooi AC, Cook M, Ghanem G, Karra Gurunath R, Keilholz U, van Kempen L, Leyvraz S, Mihm M, Newton-Bishop J, Patel P, Robert C, Schadendorf D, de Schaetzen G, Spatz A, de Vries E, Eggermont AM. EORTC Melanoma Group achievements. EJC Suppl 2012. [DOI: 10.1016/s1359-6349(12)70020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
37
|
Ulceration and stage are predictive of interferon efficacy in melanoma: Results of the phase III adjuvant trials EORTC 18952 and EORTC 18991. Eur J Cancer 2012; 48:218-25. [DOI: 10.1016/j.ejca.2011.09.028] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 09/28/2011] [Indexed: 11/20/2022]
|
38
|
Abstract
With an incidence that is increasing at 2–5% per year, cutaneous melanoma is an international scourge that disproportionately targets young individuals. Despite much research, the treatment of advanced disease is still quite challenging. Immunotherapy with high-dose interferon-α2b or interleukin-2 benefits a select group of patients in the adjuvant and metastatic settings, respectively, with significant attendant toxicity. Advances in the biology of malignant melanoma and the role of immunomodulatory therapy have produced advances that have stunned the field. In this paper, we review the data for the use of interferon-α2b in various dosing ranges, vaccine therapy, and the role of radiotherapy in the adjuvant setting for malignant melanoma. Recent trials in the metastatic setting using anticytoxic T-lymphocyte antigen-4 (anti-CTLA-4) monoclonal antibody therapy and BRAF inhibitor therapy have demonstrated clear benefit with prolongation of survival. Trials investigating combinations of these novel agents with existing immunomodulators are at present underway.
Collapse
|
39
|
Changes of ferritin and CRP levels in melanoma patients treated with adjuvant interferon-α (EORTC 18952) and prognostic value on treatment outcome. Melanoma Res 2011; 21:344-51. [PMID: 21546857 DOI: 10.1097/cmr.0b013e328346c17f] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Adjuvant therapy with interferon-α (IFN) only benefits a small subgroup of melanoma patients and a predictive marker selecting responders does not exist. IFN induces increased ferritin and decreased C-reactive protein (CRP) levels; however, an association with treatment effect was not studied. Serum was collected from patients participating in the European Organization for Research and Treatment of Cancer 18 952 trial comparing adjuvant treatment with IFN to observation. Serial ferritin and CRP levels were determined using enzyme-linked immunosorbent assays, before treatment and up to 24 months. Ferritin levels are influenced by sex and age; therefore ratios of serial ferritin and CRP values with corresponding pretreatment values were calculated. Cox regression model and landmark method at end of induction and 6 months were used to evaluate the association between ferritin, CRP and distant metastasis-free survival (DMFS). Baseline ferritin levels were comparable in the two treatment groups (P=0.92). However, ferritin ratios were significantly higher in IFN-treated patients (N=96) compared with untreated patients (N=21) at end of induction (mean: 2.88 vs. 0.75; P=0.0003) and at 6 months (mean: 3.18 vs. 1.02; P=0.009). In the IFN arm, higher ferritin ratios at end of induction and at 6 months were not associated with improved outcome (respectively, P=0.66 and 0.86). Concerning CRP ratios, no differences between the treatment groups, neither an association with DMFS, were observed. Administration of IFN in melanoma patients induced increase in ferritin levels but not in CRP levels. Ferritin and CRP ratios have no prognostic value regarding DMFS.
Collapse
|
40
|
Shada AL, Walters DM, Tierney SN, Slingluff CL. Surgical resection for bulky or recurrent axillary metastatic melanoma. J Surg Oncol 2011; 105:21-5. [PMID: 21826672 DOI: 10.1002/jso.22058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 07/18/2011] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Metastatic melanoma has few FDA approved treatments, and aggressive surgical resection has to be considered for management of bulky axillary metastases. We hypothesized that axillary resection in this setting is well tolerated and improves symptoms in the majority of patients. METHODS We reviewed a prospectively collected database and identified 47 stage IIIC and IV patients with axillary nodal disease greater than 5 cm (68%), recurrent disease (36%), or disease adherent to axillary neurovascular structures (45%). Paresthesias, pain, and bleeding were present in 40% of patients, and were stable or improved after surgery in 75%. Most patients were asymptomatic prior to resection, and underwent resection for prevention of potential symptoms. RESULTS Most patients underwent outpatient surgery. Postoperative complications included lymphedema (34%), range of motion limitation (23%), wound infection (17%), and neuropathic pain (17%). Among symptomatic patients, average time to progression was 3 months, compared to 9.5 months in asymptomatic patients (P = 0.08). Five-year survival was lower (16%) in symptomatic patients than in asymptomatic patients (35%, P = 0.001). DISCUSSION Surgery for bulky axillary melanoma metastases is well tolerated, and should be considered in the management of Stage III or IV melanoma. Resection prior to symptoms may improve quality of life and is associated with longer survival.
Collapse
Affiliation(s)
- Amber L Shada
- Department of Surgery, University of Virginia Health Sciences Center, Charlottesville, Virginia, USA
| | | | | | | |
Collapse
|
41
|
New drugs in melanoma: it's a whole new world. Eur J Cancer 2011; 47:2150-7. [PMID: 21802280 DOI: 10.1016/j.ejca.2011.06.052] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 06/23/2011] [Indexed: 01/07/2023]
Abstract
Current developments in systemic therapies for melanoma are spectacular. Over the last 40 years no one drug or combination of drugs demonstrated any impact on survival in metastatic melanoma. In contrast, in 2011 a number of new drugs will be approved. In 2011 immunomodulation with ipilimumab, a monoclonal antibody targeting the ligand CTLA-4, has been approved for patients with advanced melanoma in first- and second-line treatment by the Food and Drug Administration (FDA) and in second-line treatment by the European Medicines Agency (EMA). Also in 2011, a significant survival benefit of the combination of ipilimumab with dacarbazine compared with dacarbazine alone for first-line treatment was reported. Other monoclonal antibodies targeting T-cell ligands, such as programmed death-1 (PD-1), also show promise. Various inhibitors of v-Raf murine sarcoma viral oncogene homologue B1 (BRAF) yield high response rates in patients harbouring the BRAF-V600E mutation. A significant impact on both progression-free and overall survival was demonstrated for vemurafenib compared with dacarbazine in a phase-III trial. Approval is expected in 2011. Both drugs had only modest effects of 2-3 months on median survival, so combination therapies must be explored. BRAF inhibitors in combination with mitogen-activated protein kinase (MEK) inhibitors show great potential. Moreover, combinations of immunomodulators and pathway inhibitors are expected to be very active, and phase-III trials are planned. Pegylated interferon-α2b was approved in 2011 on the basis of the results of the European Organisation for Research and Treatment of Cancer (EORTC) 18991 phase-III trial demonstrating a sustained impact on relapse-free survival in patients with lymph-node-positive melanoma. The efficacy of adjuvant therapy with ipilimumab is assessed in the now fully accrued EORTC18071 trial. Adjuvant trials with BRAF and MEK inhibitors are in the planning phase. Never was there a more exciting period in the world of melanoma treatment.
Collapse
|
42
|
Krauze MT, Tarhini A, Gogas H, Kirkwood JM. Prognostic significance of autoimmunity during treatment of melanoma with interferon. Semin Immunopathol 2011; 33:385-91. [PMID: 21279809 PMCID: PMC8635120 DOI: 10.1007/s00281-011-0247-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/11/2011] [Indexed: 12/19/2022]
Abstract
Since the pivotal cooperative group trials in the 1980's-90's,, high-dose interferon (HDI) has been the standard of adjuvant therapy. Despite multiple other trials evaluating potential new therapies in melanoma, HDI remains the only FDA-approved therapy for stage IIB and III melanoma. Initial reports from the more recent phase III international trials of modifications of the original HDI regimen linked the appearance of autoimmunity with improved outcomes of disease. Trials of high-dose interleukin-2, many years earlier, reported anecdotal observations that were consistent with the hypothesis that autoimmunity and clinical benefit of immunotherapies of melanoma are linked with one another. The only prospectively conducted study examining the appearance of clinical and laboratory evidence of autoimmunity during HDI therapy was published by Gogas and colleagues, demonstrating statistically significant impact on relapse-free survival and overall survival. Retrospectively conducted studies of different intermediate dosage regimens of interferon (IFN) have not fully confirmed the linkage of serological evidence of autoimmunity and improved survival outcomes. With the emergence of new immunotherapies in treatment of melanoma, this review highlights the importance of autoimmunity for future applications in melanoma and reviews significant differences of past studies evaluating the appearance of autoimmunity during IFN therapy in high-risk melanoma.
Collapse
Affiliation(s)
- Michal T Krauze
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Cancer immunotherapy consists of approaches that modify the host immune system, and/or the utilization of components of the immune system, as cancer treatment. During the past 25 years, 17 immunologic products have received regulatory approval based on anticancer activity as single agents and/or in combination with chemotherapy. These include the nonspecific immune stimulants BCG and levamisole; the cytokines interferon-α and interleukin-2; the monoclonal antibodies rituximab, ofatumumab, alemtuzumab, trastuzumab, bevacizumab, cetuximab, and panitumumab; the radiolabeled antibodies Y-90 ibritumomab tiuxetan and I-131 tositumomab; the immunotoxins denileukin diftitox and gemtuzumab ozogamicin; nonmyeloablative allogeneic transplants with donor lymphocyte infusions; and the anti-prostate cancer cell-based therapy sipuleucel-T. All but two of these products are still regularly used to treat various B- and T-cell malignancies, and numerous solid tumors, including breast, lung, colorectal, prostate, melanoma, kidney, glioblastoma, bladder, and head and neck. Positive randomized trials have recently been reported for idiotype vaccines in lymphoma and a peptide vaccine in melanoma. The anti-CTLA-4 monoclonal antibody ipilumumab, which blocks regulatory T-cells, is expected to receive regulatory approval in the near future, based on a randomized trial in melanoma. As the fourth modality of cancer treatment, biotherapy/immunotherapy is an increasingly important component of the anticancer armamentarium.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Cancer Institute of Hoag Hospital , Newport Beach, California 92658, USA.
| |
Collapse
|
44
|
Leong SPL, Gershenwald JE, Soong SJ, Schadendorf D, Tarhini AA, Agarwala S, Hauschild A, Soon CWM, Daud A, Kashani-Sabet M. Cutaneous melanoma: a model to study cancer metastasis. J Surg Oncol 2011; 103:538-49. [PMID: 21480247 DOI: 10.1002/jso.21816] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nodal status in melanoma is a critically important prognostic factor for patient outcome. The survival rate drops to <10% when melanoma has spread beyond the regional lymph nodes and includes visceral involvement. In general, the process of melanoma metastasis is progressive in that dissemination of melanoma from the primary site to the regional lymph nodes occurs prior to systemic disease. The goal of this review article is to describe melanoma as a clinical model to study cancer metastasis. A future challenge is to develop a molecular taxonomy to subgroup melanoma patients at various stages of tumor progression for more accurate targeted treatment.
Collapse
Affiliation(s)
- Stanley P L Leong
- Center for Melanoma Research and Treatment and Department of Surgery, California Pacific Medical Center and Research Institute, San Francisco, California 94115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Only a subset of melanoma patients with advanced disease seems to benefit from immunotherapy. Predictive markers identifying these patients are unfortunately not available. Whether immune-related side effects could serve as predictors for treatment response or just resemble unwanted side effects from immunotherapy will be outlined in this review. RECENT FINDINGS Early studies suggested an association of immune-related side effects such as vitiligo and autoimmune thyroiditis with response in patients receiving IL-2 or IFNα. However, conflicting data have been reported as well, mentioning the effect of a higher rate of immune-related toxicities during prolonged administration of the drug in responders/survivors. This type of bias is also known as guarantee-time bias. Recently, a clearly significant and clinically relevant prolongation of survival was demonstrated in patients with metastatic melanoma treated with ipilimumab. Immune-related adverse events were associated with response to ipilimumab, however, at the cost of considerable toxicity. SUMMARY Evidence for an association of immune-related toxicities and response in patients receiving IL-2 or IFNα is weak, considering guarantee-time bias. On the contrary, this association for patients receiving anti-cytotoxic T-lymphocyte antigen-4 therapy (ipilimumab) appears much stronger. Importantly, can we uncouple tumor immunity from autoimmunity in order to optimize immunotherapy in melanoma?
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Melanoma therapy has recently seen significant progress, with several new drugs in phase II/III trials showing promising results. In this review, we discuss the most promising immunotherapies either already established or being developed, concentrating on agents for which there are high-level data to support or refute their role in treating this disease. This topic is timely, given the lengthy list of immune checkpoint inhibitors and vaccine formulations in development for melanoma. RECENT FINDINGS The discovery of immune checkpoint proteins like CTLA-4, PD-1 and CD40 and the development of antibodies and small molecules that either inhibit or promote their activity has lent a huge impetus to the immunotherapy of melanoma. The development of vaccines that include agonists of various immune signaling like the MAGE-3 ASCI has also revived the field of cancer vaccines. Melanoma is the 'poster child' for immunotherapy of cancer, since a recent randomized phase III trial showed a survival benefit for immunotherapy. SUMMARY The burgeoning field of immunotherapy for melanoma has important implications for clinicians, and for the novel paradigms of treatment and response assessment that immunotherapies will promote. The unique side-effect profile for immune checkpoint inhibitors will be a challenge but new skills for dealing with them in community based practice will be learned. The concept that physicians might see late regression, or progression followed by regression will cause a sea-change in the way patients are treated, since treating beyond progression may be suitable in some cases using immunotherapy.
Collapse
|
47
|
Shenoi MM, Shah NB, Griffin RJ, Vercellotti GM, Bischof JC. Nanoparticle preconditioning for enhanced thermal therapies in cancer. Nanomedicine (Lond) 2011; 6:545-63. [PMID: 21542691 PMCID: PMC3096482 DOI: 10.2217/nnm.10.153] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Nanoparticles show tremendous promise in the safe and effective delivery of molecular adjuvants to enhance local cancer therapy. One important form of local cancer treatment that suffers from local recurrence and distant metastases is thermal therapy. In this article, we review a new concept involving the use of nanoparticle-delivered adjuvants to 'precondition' or alter the vascular and immunological biology of the tumor to enhance its susceptibility to thermal therapy. To this end, a number of opportunities to combine nanoparticles with vascular and immunologically active agents are reviewed. One specific example of preconditioning involves a gold nanoparticle tagged with a vascular targeting agent (i.e., TNF-α). This nanoparticle embodiment demonstrates preconditioning through a dramatic reduction in tumor blood flow and induction of vascular damage, which recruits a strong and sustained inflammatory infiltrate in the tumor. The ability of this nanoparticle preconditioning to enhance subsequent heat or cold thermal therapy in a variety of tumor models is reviewed. Finally, the potential for future clinical imaging to judge the extent of preconditioning and thus the optimal timing and extent of combinatorial thermal therapy is discussed.
Collapse
Affiliation(s)
- Mithun M. Shenoi
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Neha B. Shah
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | - John C. Bischof
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Urological Surgery, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
48
|
Melanoma - The pieces of the puzzle finally start coming together! Mol Oncol 2011; 5:113-5. [DOI: 10.1016/j.molonc.2011.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 03/25/2011] [Indexed: 11/17/2022] Open
|
49
|
Current world literature. Curr Opin Oncol 2011; 23:227-34. [PMID: 21307677 DOI: 10.1097/cco.0b013e328344b687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Bouwhuis MG, ten Hagen TLM, Eggermont AMM. Immunologic functions as prognostic indicators in melanoma. Mol Oncol 2011; 5:183-9. [PMID: 21367679 DOI: 10.1016/j.molonc.2011.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 01/27/2011] [Indexed: 02/08/2023] Open
Abstract
Outcome in melanoma patients with advanced disease is poor and systemic treatment seems to benefit only a subset of patients. Predictive markers identifying these patients are currently not available. Early studies showed an association of immune-related side effects such as vitiligo and autoimmune thyroiditis with response to IL-2 or IFNα treatment. However, conflicting data have been reported as well, mentioning the effect of a higher rate of immune-related toxicities during prolonged administration of the drug in responders. The review discusses the prognostic significance of autoimmunity during various forms of immunotherapy and stresses the importance of correcting for guarantee-time bias. In addition, other immune-related factors which have been associated with melanoma prognosis such as, CRP, white blood cell count, absolute lymphocyte count and human leukocyte antigen will be reviewed as well. A better understanding of the immune system and the host-tumor interactions should ultimately lead to more effective treatment. A major challenge expected to be addressed in future is proving ways to uncouple tumor immunity from autoimmunity.
Collapse
Affiliation(s)
- Marna G Bouwhuis
- Department of Surgery, Division Surgical Oncology, Erasmus University Medical Center - Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|