201
|
Frerich B. [Treatment of cutaneous malignant melanoma in the head and neck region : An update]. HNO 2018; 66:857-873. [PMID: 30302496 DOI: 10.1007/s00106-018-0573-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Major progress has been made in the treatment of malignant melanoma during recent years. On the one hand, reliable evidence-based recommendations regarding surgical resection as well as lymph node management have been established on the basis of results from randomized multicenter studies. On the other hand, the advent of targeted therapies has led to spectacular improvements in the treatment of metastasized melanomas. Knowledge concerning treatment algorithms and updates thereof is also essential for maxillofacial surgeons. The German S3 guideline on diagnosis and treatment of melanoma was published in 2013 and revised in 2016 and 2018. This article informs readers about these developments and addresses the items relevant for treatment of head and neck melanomas in particular.
Collapse
Affiliation(s)
- B Frerich
- Klinik und Poliklinik für Mund‑, Kiefer- und Plastische Gesichtschirurgie, Universitätsmedizin Rostock, Schillingallee 35, 18057, Rostock, Deutschland.
| |
Collapse
|
202
|
Koppolu V, Rekha Vasigala VK. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J Cancer Res Ther 2018; 14:1167-1175. [PMID: 30488824 DOI: 10.4103/jcrt.jcrt_1290_16] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Clinical management of metastatic melanoma suffered historically from a lack of effective targeted and immunotherapies due to short-lived clinical responses. Recent advances in our understanding of tumor-immune signaling pathways, discovery of immunosuppressive checkpoints, and subsequent development of antibodies that target these checkpoints reverses the situation to some extent. Two antibodies ipilimumab and nivolumab gained Food and Drug administration approval for the treatment of metastatic melanoma and target two major immunosuppressive checkpoints cytotoxic T lymphocyte antigen and programmed cell death protein 1 (PD-1), respectively. Nivolumab binds to PD-1, prevents PD-1 interaction with ligand Programmed death ligand 1 (PD-L1), and thus releases the T-cell exhaustion events (such as T cell apoptosis, decrease in T cell proliferation, etc.) leading to buildup of potent tumor-specific immune response. Successful Phase I-III results with remarkable antitumor activity and safety led to approval of nivolumab against ipilimumab refractory metastatic melanoma. Nivolumab therapy is exciting in that it not only provides substantial benefit but also provides durable responses. This review focuses on the evolution of immunotherapy leading to nivolumab approval and its potential in treating melanoma either alone or in combination with other therapies.
Collapse
Affiliation(s)
- Veerendra Koppolu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Veneela Krishna Rekha Vasigala
- Department of General Medicine, Rangaraya Medical College, NTR University of Health Sciences, Vijayawada, Andhra Pradesh, India
| |
Collapse
|
203
|
Cherny NI, de Vries EGE. Improving on Tail-of-the-Curve Evaluation With the American Society of Clinical Oncology Value Framework. JAMA Oncol 2018; 4:1437. [PMID: 30128567 DOI: 10.1001/jamaoncol.2018.3264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Nathan I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | |
Collapse
|
204
|
Challenges and Opportunities of Neoadjuvant Treatment in Locally Advanced Melanoma. Am J Clin Dermatol 2018; 19:639-646. [PMID: 30039289 DOI: 10.1007/s40257-018-0371-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Locally advanced and metastatic melanoma have historically had poor survival outcomes. Long-term follow-up of both targeted therapies and immune checkpoint inhibitors has confirmed the survival benefit of these agents in stage IV melanoma, and recent studies have now demonstrated relapse-free survival benefits from these targeted and immunotherapeutic agents in the adjuvant setting. Neoadjuvant treatment of locally advanced melanoma, including in-transit disease, is now under investigation. Clinical trials have shown early promising results using either combination targeted therapy or immune checkpoint inhibitors. Neoadjuvant treatment may improve surgical morbidity, but balancing treatment efficacy and toxicity has already been challenging in the use of combination immune checkpoint inhibitors preoperatively. While improvement in relapse-free survival has been noted, additional follow-up of patients receiving neoadjuvant treatment will be necessary to report on long-term outcomes. Neoadjuvant treatment also provides additional translational research opportunities to determine predictive biomarkers for targeted therapy and immune checkpoint inhibitors. Evidence of early resistance to treatment may also lead to novel combination therapies to explore in future clinical trials. While neoadjuvant treatment in locally advanced melanoma has exciting potential, more investigation is necessary to determine efficacious regimens with manageable toxicities.
Collapse
|
205
|
Ibrahim T, Mateus C, Baz M, Robert C. Older melanoma patients aged 75 and above retain responsiveness to anti-PD1 therapy: results of a retrospective single-institution cohort study. Cancer Immunol Immunother 2018; 67:1571-1578. [PMID: 30056599 PMCID: PMC11028036 DOI: 10.1007/s00262-018-2219-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 07/25/2018] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The utility of immunotherapy in elderly melanoma patients is debated. We aimed in this study to evaluate the efficacy and tolerability of immunotherapy among elderly patients. METHOD This is a retrospective single-institution cohort study. Patients aged 75 years and above who had been treated with nivolumab, pembrolizumab or ipilimumab for advanced or metastatic melanoma, were included. Patients and disease characteristics were collected using electronic medical records. Objective response was determined according to the immune-related response criteria. Drug-related toxicities (DRT) were graded according to the CTCAE v4.03. RESULTS 99 patients were included with a mean age of 80 years (SD = 4). One patient received nivolumab and ipilimumab combination, but died because of drug-related diverticulitis. Median PFS on pembrolizumab, nivolumab or ipilimumab were equal to 11.9 (95% CI 5.4-18.4), 1.4 (95% CI 0.01-2.8), and 2.8 months (95% CI 2.6-3), respectively, while objective response rates were equal to 51.6, 12.5, and 17.3%, respectively. Median OS was not reached in patients who received only pembrolizumab, 8.7 months in the ipilimumab only group, and 23 months in patients receiving several immune therapies sequentially. Pembrolizumab, nivolumab, and ipilimumab grade 3-4 DRT rates were equal to 24.2, 62.5, and 32.7% respectively, while discontinuation rates were equal to 43.5, 62.5, and 28.8%, respectively. CONCLUSIONS Our study suggests that immunotherapy is effective and well tolerated in the elderly. The PFS on pembrolizumab was greater than expected, a finding that needs to be investigated further.
Collapse
Affiliation(s)
- Tony Ibrahim
- Department of Medical Oncology, Gustave Roussy Institut, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
| | - Christine Mateus
- Dermatology Department, Gustave Roussy Institut, Villejuif, France
| | - Maria Baz
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Caroline Robert
- Dermatology Department, Gustave Roussy Institut, Villejuif, France
| |
Collapse
|
206
|
Varn FS, Wang Y, Cheng C. A B cell-derived gene expression signature associates with an immunologically active tumor microenvironment and response to immune checkpoint blockade therapy. Oncoimmunology 2018; 8:e1513440. [PMID: 30546953 PMCID: PMC6287894 DOI: 10.1080/2162402x.2018.1513440] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors have shown great potential in treating solid tumors, inducing durable remission and prolonged survival time in responders. Despite their promise, a large fraction of patients remains unresponsive to these treatments highlighting the need for biomarkers that can predict patient sensitivity. Pre-treatment gene expression profiles for patients receiving immune checkpoint inhibitors have recently become available, establishing a new medium by which to discover biomarkers that predict therapy response. In this study, we mined for transcriptomic correlates of response by applying immune cell-derived gene expression signatures to publicly available datasets containing matched gene expression and response efficacy information. These datasets were comprised of urothelial carcinoma patients receiving anti-PD-L1 (n = 25), melanoma patients receiving anti-PD-1 (n = 28), and melanoma patients receiving anti-CTLA-4 (n = 42). We identified one signature, derived from a subpopulation of B cells, with scores that were significantly and reproducibly elevated in patients experiencing clinical benefit following therapy targeting the PD-1/PD-L1 axis and were additionally elevated in patients responsive to anti-CTLA-4 therapy. Multivariate models revealed that this signature was associated with response independent of other response-predictive biomarkers, including tumor mutation burden. Functional annotation of the signature revealed it to be associated with features indicative of an immunologically active microenvironment, including B and T cell activation as well as antigen presentation activity. The preliminary findings presented detail a transcriptomic signature associated with response to multiple checkpoint inhibitors and suggest novel biological associations that warrant further investigation.
Collapse
Affiliation(s)
- Frederick S Varn
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Yue Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Chao Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Norris Cotton Cancer Center, One Medical Center Drive, Lebanon, NH, USA.,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, USA
| |
Collapse
|
207
|
Valnet-Rabier MB, Marcucci C, Limat S, Davani S, Aubin F, Nerich V. [Acceptability and effectiveness of immunotherapy in patients with melanoma]. Therapie 2018; 74:355-367. [PMID: 30193804 DOI: 10.1016/j.therap.2018.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 11/26/2022]
Abstract
The immunotherapies known as "inhibitors of checkpoint" (ICP) are monoclonal antibodies used since 2010 and have dramatically modified the management of the advanced or metastatic melanomas. By reactivating the anti-tumoral immune response, these antibodies can activate the immune system in all the tissues with a risk to induce immune related adverse events (IrAE). Thus, the adverse effect's profile of ICP is considered as very different from that usually associated with conventional chemotherapies. The objectives of our retrospective monocentric study were the evaluation of the real life's safety and efficiency of the ipilimumab and the pembrolizumab in patients with an advanced melanoma. Seventy-two patients treated by ipilimumab and\or pembrolizumab between August 1st, 2008 and December 31st, 2016 were investigated. The main IrAE occurring involved the gastro- intestinal, skin, and the endocrine systems. The average onset time of IrAE was 39, 104 and 68 days, respectively and their respective duration was of 67, 50 and 111 days. There were 13 events of grade III and IV along with one death. The overall survival was 5 months for the patients treated in monotherapy with ipilimumab, and 14 months for those treated by pembrolizumab. Our real life's study tends to confirm the current safety profile of ICP treatment. Moreover and according to our analyses, the drug sequence seems to have a global survival impact.
Collapse
Affiliation(s)
| | | | - Samuel Limat
- Pôle pharmaceutique, CHU de Besançon, 25030 Besançon, France; Inserm, EFS BFC, UMR1098, interactions hôte-greffon-tumeur/ingénierie cellulaire et génique, université Bourgogne Franche-Comté, 25000 Besançon, France
| | - Siamak Davani
- Centre régional de pharmacovigilance, CHU de Besançon, 25030 Besançon, France
| | - François Aubin
- Service de dermatologie, CHU de Besançon, 25030 Besançon, France
| | - Virginie Nerich
- Pôle pharmaceutique, CHU de Besançon, 25030 Besançon, France; Inserm, EFS BFC, UMR1098, interactions hôte-greffon-tumeur/ingénierie cellulaire et génique, université Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
208
|
Hansen ED, Wang X, Case AA, Puzanov I, Smith T. Immune Checkpoint Inhibitor Toxicity Review for the Palliative Care Clinician. J Pain Symptom Manage 2018; 56:460-472. [PMID: 29792979 DOI: 10.1016/j.jpainsymman.2018.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have opened an exciting chapter in the treatment of patients with advanced cancer. For the palliative care clinician, however, ICIs present several new challenges, including new ways to define treatment success, as well as treatment-related toxicities that differ in nature and timing from traditional chemotherapy. In this article, we review the mechanism of action of ICIs, as well as selected published data supporting the efficacy of ICIs in patients with advanced cancer. In addition, we summarize existing data of ICI toxicity prevalence, patterns of severity, and timing of onset. Finally, we briefly review key principles from published guidelines on the management of ICI toxicities.
Collapse
Affiliation(s)
- Eric D Hansen
- Department of Supportive Care, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| | - Xiao Wang
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Amy A Case
- Department of Supportive Care, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tom Smith
- Palliative Medicine Program, Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
209
|
Calabrò L, Morra A, Cornelissen R, Aerts J, Maio M. Immune checkpoint blockade therapy of mesothelioma: a clinical and radiological challenge. Cancer Immunol Immunother 2018; 67:1317-1324. [PMID: 29943157 PMCID: PMC11028073 DOI: 10.1007/s00262-018-2191-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 06/22/2018] [Indexed: 02/08/2023]
Abstract
Treatment of malignant pleural mesothelioma (MPM) represents a highly unmet medical need. Here, we discuss the results and therapeutic potential of first- and second-generation immunomodulatory antibodies targeting distinct immune checkpoints for the treatment of MPM, as well as their prospective therapeutic role in combination strategies. We also discuss the role of appropriate radiological criteria of response for MPM and the potential need of ad hoc criteria of disease evaluation in MPM patients undergoing treatment with immunotherapeutic agents.
Collapse
Affiliation(s)
- Luana Calabrò
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Viale Mario Bracci n. 16, 53100, Siena, Italy.
| | - Aldo Morra
- Department of Radiology, Euganea Medica Diagnostic Center, Padua, Italy
| | | | - Joachim Aerts
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michele Maio
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Viale Mario Bracci n. 16, 53100, Siena, Italy
| |
Collapse
|
210
|
Chen P, Chen F, Zhou B. Comparisons of therapeutic efficacy and safety of ipilimumab plus GM-CSF versus ipilimumab alone in patients with cancer: a meta-analysis of outcomes. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2025-2038. [PMID: 30013322 PMCID: PMC6037403 DOI: 10.2147/dddt.s154258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Recent clinical studies have shown that initial therapy with combined cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) blockade and granulocyte-macrophage colony-stimulating factor (GM-CSF)-based immunotherapies can enhance the antitumor efficacy of this approach. A key unanswered question is whether systemic GM-CSF enhances CTLA-4 blockade. Thus, the objective of this study was taking a meta-analysis of randomized controlled trials to compare the effect of ipilimumab plus GM-CSF versus ipilimumab alone on overall response, overall survival, and progression-free survival, as well as the risk of adverse events (AEs) in patients with cancer. Materials and methods Searches were made in electronic databases PubMed and Embase, and conference abstracts published by the American Society of Clinical Oncology from 2000 to 2017. Statistical analyses were carried out using either random-effects or fixed-effects models according to the heterogeneity of eligible studies. Results Six trials comprising of 445 patients were included in the meta-analysis. Combination group was superior to the ipilimumab alone in overall response rate, progression-free survival, and overall survival rate (combined relative risk [RR]=1.34, 95% CI: 1.24–1.45, P=0.09; combined hazard ratio [HR]=0.57, 95% CI: 0.32–1.02, P=0.06; combined HR=0.70, 95% CI: 0.60–0.82, P<0.001). Patients with combination therapies had a lower incidence of AEs including high-grade diarrhea (combined RR=0.27, 95% CI: 0.11–0.70, P=0.007), nausea (combined RR=0.25, 95% CI: 0.07–0.89, P=0.03), colitis (combined RR=0.34, 95% CI: 0.13–0.86, P=0.02), and fatigue (combined RR=0.91, 95% CI: 0.37–2.2.3, P=0.84) compared to the group having ipilimumab alone. Conclusion These data suggested that the combination of ipilimumab and GM-CSF was associated with a significant improvement in overall survival and lower high-grade toxicities, but there is no difference in overall response rate and progression-free survival among the cancer patients. Therefore, large-scale and well-designed studies are needed to summarize and analyze the data to draw a more convincing conclusion.
Collapse
Affiliation(s)
- Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China,
| | - Fuchao Chen
- Department of Pharmacy, Dongfeng Hospital, Hubei University of Medicine, Shiyan, Hubei, 442008, People's Republic of China
| | - Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China, .,School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China,
| |
Collapse
|
211
|
Hanna TP, Nguyen P, Baetz T, Booth CM, Eisenhauer E. A Population-based Study of Survival Impact of New Targeted and Immune-based Therapies for Metastatic or Unresectable Melanoma. Clin Oncol (R Coll Radiol) 2018; 30:609-617. [PMID: 30196844 DOI: 10.1016/j.clon.2018.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 11/16/2022]
Abstract
AIMS New targeted drugs and immune therapies reported since 2010 for metastatic or unresectable melanoma (MM) have shown improved survival in randomised trials. We studied the uptake of these new drugs and their impact on population-based survival. MATERIALS AND METHODS This was a retrospective, population-based cohort study of all patients treated for MM in Ontario 2007-2015. Provincial administrative sources covering the whole population identified palliative systemic therapy, radiotherapy and metastasis surgery. Temporal trends in utilisation and survival were investigated, as was survival of treatments predefined as 'new drugs' (BRAF or MEK inhibitors, anti-CTLA4 and anti-PD-1 antibodies). RESULTS We identified 2793 treated MM patients. First treatment was systemic therapy (46%), radiotherapy (41%) and metastasis surgery (14%). Systemic treatment increased from 53% of patients (2007) to 75% (2015). New drug treatments increased from <6% of known first-line regimens in 2007 to 82% in 2015. One and 2 year overall survival was 28% and 15%, respectively, for all MM 2007-2009, rising to 46% and 35% for 2014-2015 (adjusted hazard ratio 0.56, 95% confidence interval 0.49-0.63, P < 0.0001). Survival gains were observed primarily among those cases initially treated with systemic therapy, which became dominated by the use of new drugs over the study period (2 year overall survival 16% 2007-2009 versus 44% 2014-2015; adjusted hazard ratio 0.46, 95% confidence interval 0.38-0.56, P < 0.0001). CONCLUSIONS Utilisation of new targeted drugs and immune therapies for MM has increased considerably in routine practice 2007-2015. Consistent with the results of clinical trials, adoption was associated with substantial increases in survival of patients in the general population.
Collapse
Affiliation(s)
- T P Hanna
- Division of Cancer Care and Epidemiology, Cancer Research Institute at Queen's University, Kingston, Ontario, Canada; Department of Oncology, Queen's University, Kingston, Ontario, Canada; Institute for Clinical Evaluative Sciences at Queen's University, Kingston, Ontario, Canada.
| | - P Nguyen
- Institute for Clinical Evaluative Sciences at Queen's University, Kingston, Ontario, Canada
| | - T Baetz
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - C M Booth
- Division of Cancer Care and Epidemiology, Cancer Research Institute at Queen's University, Kingston, Ontario, Canada; Department of Oncology, Queen's University, Kingston, Ontario, Canada; Institute for Clinical Evaluative Sciences at Queen's University, Kingston, Ontario, Canada
| | - E Eisenhauer
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
212
|
Anagnostou V, Yarchoan M, Hansen AR, Wang H, Verde F, Sharon E, Collyar D, Chow LQM, Forde PM. Immuno-oncology Trial Endpoints: Capturing Clinically Meaningful Activity. Clin Cancer Res 2018; 23:4959-4969. [PMID: 28864724 DOI: 10.1158/1078-0432.ccr-16-3065] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/26/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022]
Abstract
Immuno-oncology (I-O) has required a shift in the established paradigm of toxicity and response assessment in clinical research. The design and interpretation of cancer clinical trials has been primarily driven by conventional toxicity and efficacy patterns observed with chemotherapy and targeted agents, which are insufficient to fully inform clinical trial design and guide therapeutic decisions in I-O. Responses to immune-targeted agents follow nonlinear dose-response and dose-toxicity kinetics mandating the development of novel response evaluation criteria. Biomarker-driven surrogate endpoints may better capture the mechanism of action and biological response to I-O agents and could be incorporated prospectively in early-phase I-O clinical trials. While overall survival remains the gold standard for evaluation of clinical efficacy of I-O agents in late-phase clinical trials, exploration of potential novel surrogate endpoints such as objective response rate and milestone survival is to be encouraged. Patient-reported outcomes should also be assessed to help redefine endpoints for I-O clinical trials and drive more efficient drug development. This paper discusses endpoints used in I-O trials to date and potential optimal endpoints for future early- and late-phase clinical development of I-O therapies.
Collapse
Affiliation(s)
- Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Yarchoan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre-University Health Network, Department of Medicine, University of Toronto, Toronto, Canada
| | - Hao Wang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Franco Verde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | | | | | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
213
|
Sullivan RJ, Atkins MB, Kirkwood JM, Agarwala SS, Clark JI, Ernstoff MS, Fecher L, Gajewski TF, Gastman B, Lawson DH, Lutzky J, McDermott DF, Margolin KA, Mehnert JM, Pavlick AC, Richards JM, Rubin KM, Sharfman W, Silverstein S, Slingluff CL, Sondak VK, Tarhini AA, Thompson JA, Urba WJ, White RL, Whitman ED, Hodi FS, Kaufman HL. An update on the Society for Immunotherapy of Cancer consensus statement on tumor immunotherapy for the treatment of cutaneous melanoma: version 2.0. J Immunother Cancer 2018; 6:44. [PMID: 29848375 PMCID: PMC5977556 DOI: 10.1186/s40425-018-0362-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/17/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer immunotherapy has been firmly established as a standard of care for patients with advanced and metastatic melanoma. Therapeutic outcomes in clinical trials have resulted in the approval of 11 new drugs and/or combination regimens for patients with melanoma. However, prospective data to support evidence-based clinical decisions with respect to the optimal schedule and sequencing of immunotherapy and targeted agents, how best to manage emerging toxicities and when to stop treatment are not yet available. METHODS To address this knowledge gap, the Society for Immunotherapy of Cancer (SITC) Melanoma Task Force developed a process for consensus recommendations for physicians treating patients with melanoma integrating evidence-based data, where available, with best expert consensus opinion. The initial consensus statement was published in 2013, and version 2.0 of this report is an update based on a recent meeting of the Task Force and extensive subsequent discussions on new agents, contemporary peer-reviewed literature and emerging clinical data. The Academy of Medicine (formerly Institute of Medicine) clinical practice guidelines were used as a basis for consensus development with an updated literature search for important studies published between 1992 and 2017 and supplemented, as appropriate, by recommendations from Task Force participants. RESULTS The Task Force considered patients with stage II-IV melanoma and here provide consensus recommendations for how they would incorporate the many immunotherapy options into clinical pathways for patients with cutaneous melanoma. CONCLUSION These clinical guidleines provide physicians and healthcare providers with consensus recommendations for managing melanoma patients electing treatment with tumor immunotherapy.
Collapse
Affiliation(s)
- Ryan J. Sullivan
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | | | | | - Sanjiv S. Agarwala
- St. Luke’s Cancer Center and Temple University, Center Valley, PA 18034 USA
| | | | | | | | | | | | | | - Jose Lutzky
- Mt. Sinai Medical Center, Miami Beach, FL 33140 USA
| | | | | | | | - Anna C. Pavlick
- New York University Cancer Institute, New York, NY 10016 USA
| | | | - Krista M. Rubin
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - William Sharfman
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231 USA
| | | | | | - Vernon K. Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612 USA
| | | | | | - Walter J. Urba
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR 97213 USA
| | | | | | | | - Howard L. Kaufman
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| |
Collapse
|
214
|
Cherny NI, Dafni U, Bogaerts J, Latino NJ, Pentheroudakis G, Douillard JY, Tabernero J, Zielinski C, Piccart MJ, de Vries EGE. ESMO-Magnitude of Clinical Benefit Scale version 1.1. Ann Oncol 2018; 28:2340-2366. [PMID: 28945867 DOI: 10.1093/annonc/mdx310] [Citation(s) in RCA: 481] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background The ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS) version 1.0 (v1.0) was published in May 2015 and was the first version of a validated and reproducible tool to assess the magnitude of clinical benefit from new cancer therapies. The ESMO-MCBS was designed to be a dynamic tool with planned revisions and updates based upon recognition of expanding needs and shortcomings identified since the last review. Methods The revision process for the ESMO-MCBS incorporates a nine-step process: Careful review of critiques and suggestions, and identification of problems in the application of v1.0; Identification of shortcomings for revision in the upcoming version; Proposal and evaluation of solutions to address identified shortcomings; Field testing of solutions; Preparation of a near-final revised version for peer review for reasonableness by members of the ESMO Faculty and Guidelines Committee; Amendments based on peer review for reasonableness; Near-final review by members of the ESMO-MCBS Working Group and the ESMO Executive Board; Final amendments; Final review and approval by members of the ESMO-MCBS Working Group and the ESMO Executive Board. Results Twelve issues for revision or amendment were proposed for consideration; proposed amendments were formulated for eight identified shortcomings. The proposed amendments are classified as either structural, technical, immunotherapy triggered or nuanced. All amendments were field tested in a wide range of studies comparing scores generated with ESMO-MCBS v1.0 and version 1.1 (v1.1). Conclusions ESMO-MCBS v1.1 incorporates 10 revisions and will allow for scoring of single-arm studies. Scoring remains very stable; revisions in v1.1 alter the scores of only 12 out of 118 comparative studies and facilitate scoring for single-arm studies.
Collapse
Affiliation(s)
- N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel;.
| | - U Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens & Frontier Science Foundation-Hellas, Athens, Greece
| | - J Bogaerts
- Methodology Direction, EORTC Headquarters, Brussels, Belgium
| | | | - G Pentheroudakis
- Medical Oncology Department, Ioannina University Hospital, Ioannina, Greece
| | | | - J Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - C Zielinski
- Division of Oncology, Medical University Vienna, Vienna, Austria
| | - M J Piccart
- Jules Bordet Institute, Universite Libre de Bruxelles, Brussels, Belgium
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
215
|
Hogan SA, Levesque MP, Cheng PF. Melanoma Immunotherapy: Next-Generation Biomarkers. Front Oncol 2018; 8:178. [PMID: 29896449 PMCID: PMC5986946 DOI: 10.3389/fonc.2018.00178] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 01/05/2023] Open
Abstract
The recent emergence of cancer immunotherapies initiated a significant shift in the clinical management of metastatic melanoma. Prior to 2011, melanoma patients only had palliative treatment solutions which offered little to no survival benefit. In 2018, with immunotherapy, melanoma patients can now contemplate durable or even complete remission. Treatment with novel immune checkpoint inhibitors, anti-cytotoxic T-lymphocyte protein 4 and anti-programmed cell death protein 1, clearly result in superior median and long-term survivals compared to standard chemotherapy; however, more than half of the patients do not respond to immune checkpoint blockade. Currently, clinicians do not have any effective way to stratify melanoma patients for immunotherapies. Research is now focusing on identifying biomarkers which could predict a patient’s response prior treatment initiation (or very early during treatment course), in order to maximize therapeutic efficacy, avoid unnecessary costs, and undesirable heavy side effects for the patient. Given the rapid developments in this field and the translational potential for some of the biomarkers, we will summarize the current state of biomarker research for immunotherapy in melanoma, with an emphasis on omics technologies such as next-generation sequencing and mass cytometry (CyTOF).
Collapse
Affiliation(s)
- Sabrina A Hogan
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| | - Phil F Cheng
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| |
Collapse
|
216
|
Figueiredo CR, Azevedo RA, Mousdell S, Resende-Lara PT, Ireland L, Santos A, Girola N, Cunha RLOR, Schmid MC, Polonelli L, Travassos LR, Mielgo A. Blockade of MIF-CD74 Signalling on Macrophages and Dendritic Cells Restores the Antitumour Immune Response Against Metastatic Melanoma. Front Immunol 2018; 9:1132. [PMID: 29875777 PMCID: PMC5974174 DOI: 10.3389/fimmu.2018.01132] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/04/2018] [Indexed: 12/15/2022] Open
Abstract
Mounting an effective immune response against cancer requires the activation of innate and adaptive immune cells. Metastatic melanoma is the most aggressive form of skin cancer. While immunotherapies have shown a remarkable success in melanoma treatment, patients develop resistance by mechanisms that include the establishment of an immune suppressive tumor microenvironment. Thus, understanding how metastatic melanoma cells suppress the immune system is vital to develop effective immunotherapies against this disease. In this study, we find that macrophages (MOs) and dendritic cells (DCs) are suppressed in metastatic melanoma and that the Ig-CDR-based peptide C36L1 is able to restore MOs and DCs' antitumorigenic and immunogenic functions and to inhibit metastatic growth in lungs. Specifically, C36L1 treatment is able to repolarize M2-like immunosuppressive MOs into M1-like antitumorigenic MOs, and increase the number of immunogenic DCs, and activated cytotoxic T cells, while reducing the number of regulatory T cells and monocytic myeloid-derived suppressor cells in metastatic lungs. Mechanistically, we find that C36L1 directly binds to the MIF receptor CD74 which is expressed on MOs and DCs, disturbing CD74 structural dynamics and inhibiting MIF signaling on these cells. Interfering with MIF-CD74 signaling on MOs and DCs leads to a decrease in the expression of immunosuppressive factors from MOs and an increase in the capacity of DCs to activate cytotoxic T cells. Our findings suggest that interfering with MIF-CD74 immunosuppressive signaling in MOs and DCs, using peptide-based immunotherapy can restore the antitumor immune response in metastatic melanoma. Our study provides the rationale for further development of peptide-based therapies to restore the antitumor immune response in metastatic melanoma.
Collapse
Affiliation(s)
- Carlos R. Figueiredo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ricardo A. Azevedo
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sasha Mousdell
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Pedro T. Resende-Lara
- Laboratory of Computational Biology and Bioinformatics, Federal University of ABC, Santo André, Brazil
- Laboratoire de Biologie et Pharmacologie Appliquées (LBPA), UMR 8113, Ecole Normale Supérieure, Cachan, France
| | - Lucy Ireland
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Almudena Santos
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Natalia Girola
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Rodrigo L. O. R. Cunha
- Chemical Biology Laboratory, Natural and Human Sciences Center, Federal University of ABC, Santo André, Brazil
| | - Michael C. Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Luciano Polonelli
- Unit of Biomedical, Biotechnological and Translational Sciences, Department of Medicine and Surgery, Universitá degli Studi di Parma, Parma, Italy
| | - Luiz R. Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
217
|
De Santo C, Cheng P, Beggs A, Egan S, Bessudo A, Mussai F. Metabolic therapy with PEG-arginase induces a sustained complete remission in immunotherapy-resistant melanoma. J Hematol Oncol 2018; 11:68. [PMID: 29776373 PMCID: PMC5960181 DOI: 10.1186/s13045-018-0612-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metastatic melanoma is an aggressive skin cancer with a poor prognosis. Current treatment strategies for high-stage melanoma are based around the use of immunotherapy with immune checkpoint inhibitors such as anti-PDL1 or anti-CTLA4 antibodies to stimulate anti-cancer T cell responses, yet a number of patients will relapse and die of disease. Here, we report the first sustained complete remission in a patient with metastatic melanoma who failed two immunotherapy strategies, by targeting tumour arginine metabolism. CASE PRESENTATION A 65-year-old patient with metastatic melanoma who progressed through two immunotherapy strategies with immune checkpoint inhibitor antibodies was enrolled in a phase I study (NCT02285101) and treated with 2 mg/kg intravenously, weekly pegylated recombinant arginase (BCT-100). The patient experienced no toxicities > grade 2 and entered a complete remission which is sustained for over 30 months. RNA-sequencing identified a number of transcriptomic pathway alterations compared to control samples. The tumour had absent expression of the recycling enzymes argininosuccinate synthetase (ASS) and ornithine transcarbamylase (OTC) indicating a state of arginine auxotrophy, which was reconfirmed by immunohistochemistry, and validation in a larger cohort of melanoma tumour samples. CONCLUSIONS Targeting arginine metabolism with therapeutic arginase in arginine auxotrophic melanoma can be an effective salvage for the treatment of patients who fail immunotherapy.
Collapse
Affiliation(s)
- Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Paul Cheng
- Bio-cancer Treatment International, Ltd., Shatin, Hong Kong
| | - Andrew Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Sharon Egan
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Sutton Bonington, UK
| | | | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
218
|
Puza CJ, Cardones AR, Mosca PJ. Examining the Incidence and Presentation of Melanoma in the Cardiothoracic Transplant Population. JAMA Dermatol 2018; 154:589-591. [PMID: 29453871 PMCID: PMC5876892 DOI: 10.1001/jamadermatol.2018.0130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/18/2018] [Indexed: 02/05/2023]
Abstract
Importance The immunosuppression vital to maintaining transplanted organs comes with an increased incidence of cutaneous neoplasms. Understanding the genesis of malignant melanoma (MM) in transplant subpopulations is necessary for adequate disease surveillance. Objective To determine the incidence and timing of presentation of MM in the cardiothoracic (heart and/or lung) transplant (CTT) population. Design, Setting, and Participants This was a retrospective cohort study of 1164 patients who underwent a CTT from 2001 through 2016 with a median follow-up time of 4.3 years. The study was performed at a single academic, tertiary referral center. The retrospective database was used to identify 1164 patients who underwent a CTT at Duke University Hospital from 2001 to 2016. Ten patients were excluded from the study owing to a history of MM, resulting in 1154 patients in the study. Five patients who developed MM after CTT were identified. Exposures Exposures included tacrolimus, prednisone, and mycophenolate mofetil. Main Outcomes and Measures The primary outcome measurement was the MM incidence. Secondary outcomes included time to diagnosis and survival. Results Five of 1154 patients who underwent a CTT (0.4%) developed biopsy-proven MM at a median follow-up time of 4.3 years after transplantation at a median age of 64.5 years (range, 31.0-74.0 years). Of the 1154 patients, 923 (80%) were men. Their mean (SD) age range was 63.8 years (27.2-68.2 years). Four patients (80%) presented with stage I disease while 1 (20%) presented with stage IV disease at a median time of 2.5 years (range, 0.1-5.3 years) after transplant compared with a median time of 6.2 years (range, 0.9-8.7 years) in Duke University's renal transplant population at a median follow-up time of 6.6 years. Two patients died after transplant, 1 owing to complications of the transplant and 1 owing to metastatic MM. Conclusions and Relevance Representing one of the largest reported studies of patients with CTT developing MM, our findings suggest that the CTT population experiences an incidence of MM similar to that of other solid organ transplant recipients and with a median of 2.5 years from transplant to melanoma diagnosis. While the small scale of our study prevents far-reaching conclusions, further study is warranted to better understand the incidence, timing, and clinical ramifications of melanomagenesis in the CTT population.
Collapse
Affiliation(s)
| | - Adela R. Cardones
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina
| | - Paul J. Mosca
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
219
|
Dragomir M, Chen B, Fu X, Calin GA. Key questions about the checkpoint blockade-are microRNAs an answer? Cancer Biol Med 2018; 15:103-115. [PMID: 29951335 PMCID: PMC5994554 DOI: 10.20892/j.issn.2095-3941.2018.0006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/20/2018] [Indexed: 12/18/2022] Open
Abstract
The introduction of immune-checkpoint blockade in the cancer therapy led to a paradigm change of the management of late stage cancers. There are already multiple FDA approved checkpoint inhibitors and many other agents are undergoing phase 2 and early phase 3 clinical trials. The therapeutic indication of immune checkpoint inhibitors expanded in the last years, but still remains unclear who can benefit. MicroRNAs are small RNAs with no coding potential. By complementary pairing to the 3' untranslated region of messenger RNA, microRNAs exert posttranscriptional control of protein expression. A network of microRNAs directly and indirectly controls the expression of checkpoint receptors and several microRNAs can target multiple checkpoint molecules, mimicking the therapeutic effect of a combined immune checkpoint blockade. In this review, we will describe the microRNAs that control the expression of immune checkpoints and we will present four specific issues of the immune checkpoint therapy in cancer: (1) imprecise therapeutic indication, (2) difficult response evaluation, (3) numerous immunologic adverse-events, and (4) the absence of response to immune therapy. Finally, we propose microRNAs as possible solutions for these pitfalls. We consider that in the near future microRNAs could become important therapeutic partners of the immune checkpoint therapy.
Collapse
Affiliation(s)
- Mihnea Dragomir
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania
- Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest 4192910, Romania
| | - Baoqing Chen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Xiao Fu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - George A. Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
220
|
Linck RDM, Costa RLDP, Garicochea B. Cancer immunology and melanoma immunotherapy. An Bras Dermatol 2018; 92:830-835. [PMID: 29364441 PMCID: PMC5786399 DOI: 10.1590/abd1806-4841.201756511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/11/2017] [Indexed: 01/21/2023] Open
Abstract
The stimulation of the immune system, in order to generate an attack against
cancer cells, similarly to that which occurs in infectious disease, has long
been matter of interest in oncology; however, only limited success has been
achieved, with different treatment strategies tested in recent years. The
development of new immune checkpoint inhibitors is currently changing this
scenario, and immunotherapy is becoming a real choice among traditional
cytotoxic treatments to fight cancer. Recent reports have shown efficacy and
safety with the use of pembrolizumab, nivolumab, and ipilimumab for the
treatment of different neoplasms, especially melanoma. In this article, we
propose a review of the mechanisms of action involved in cancer immunology, the
response evaluation of immunotherapies, and its toxicity profile, as well as a
summary of the main clinical trials that led to the adoption of these new drugs
for melanoma treatment.
Collapse
|
221
|
Wieder T, Eigentler T, Brenner E, Röcken M. Immune checkpoint blockade therapy. J Allergy Clin Immunol 2018; 142:1403-1414. [PMID: 29596939 DOI: 10.1016/j.jaci.2018.02.042] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/09/2018] [Accepted: 02/27/2018] [Indexed: 12/30/2022]
Abstract
Immune checkpoints are accessory molecules that either promote or inhibit T-cell activation. Two inhibitory molecules, cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), got high attention, as inhibition of CTLA-4 or PD-1 signaling provides the first immune therapy that significantly improves the survival of patients with metastatic solid cancers. Inhibition of CTLA-4 or PD-1 was first studied in and approved for patients with metastatic melanoma. Blocking immune checkpoints is also efficient in non-small-cell lung cancer, renal cell cancers, hypermutated gastrointestinal cancers, and others. Immune responses, whether directed against infections or against tumors, are divided into 2 phases: an initiation phase and an activation phase, where the immune system recognizes a danger signal and becomes activated by innate signals to fight the danger. This reaction is fundamental for the control of infections and cancer, but needs to be turned off once the danger is controlled, because persistence of this activation ultimately causes severe tissue damage. Therefore, each activation of the immune system is followed by a termination phase, where endogenous immune suppressor molecules arrest immune responses to prevent harmful damage. In the case of cancer immune therapies, therapeutic approaches classically enhanced the initiation and activation of immune responses to increase the emergence and the efficacy of cytotoxic T lymphocytes (CTL) against cancers. In sharp contrast, immune checkpoint blockade focuses on the termination of immune responses by inhibiting immune suppressor molecules. It thus prevents the termination of immune responses or even awakes those CTLs that became exhausted during an immune response. Therefore, blocking negatively regulating immune checkpoints restores the capacity of exhausted CTL to kill the cancer they infiltrate. In addition, they drive surviving cancer cells into a still poorly defined state of dormancy. As the therapy also awakes self-reactive CTL, one downside of the therapy is the induction of organ-specific autoimmune diseases. The second downside is the exorbitant drug price that withdraws patients in need from a therapy that was developed by academic research, which impairs further academic treatment development and financially charges the public health system.
Collapse
Affiliation(s)
- Thomas Wieder
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Ellen Brenner
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
222
|
Abstract
PURPOSE OF REVIEW Prognosis of patients with advanced melanoma is dismal with a median overall survival of about 8 months and 5-year overall survival from a diagnosis of metastatic disease of roughly 10%. However, immune checkpoint inhibitors have brought indispensable benefits to melanoma patients. Here we will review the recent clinical efficacy and adverse events of immune checkpoint inhibitors for melanoma patients. RECENT FINDINGS The immune checkpoint inhibitors increase confirmed objective response and prolong progression-free and overall survival of the afflicted patients in association with maintaining their quality of life. Although diverse immune-related adverse events occur, most of them are manageable by appropriate immunomodulating agents. Clinical efficacy of immune checkpoint inhibitors continues even after discontinuation of drugs. Compared with conventional therapeutic options, the immune checkpoint inhibitors appear to prolong the survival of patients with advanced melanoma. Further clinical trials are warranted to determine whether their combinatory use with other treatment options may augment benefits or not.
Collapse
|
223
|
Bryant CE, Sutherland S, Kong B, Papadimitrious MS, Fromm PD, Hart DNJ. Dendritic cells as cancer therapeutics. Semin Cell Dev Biol 2018; 86:77-88. [PMID: 29454038 DOI: 10.1016/j.semcdb.2018.02.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/14/2017] [Accepted: 02/10/2018] [Indexed: 02/06/2023]
Abstract
The ability of immune therapies to control cancer has recently generated intense interest. This therapeutic outcome is reliant on T cell recognition of tumour cells. The natural function of dendritic cells (DC) is to generate adaptive responses, by presenting antigen to T cells, hence they are a logical target to generate specific anti-tumour immunity. Our understanding of the biology of DC is expanding, and they are now known to be a family of related subsets with variable features and function. Most clinical experience to date with DC vaccination has been using monocyte-derived DC vaccines. There is now growing experience with alternative blood-derived DC derived vaccines, as well as with multiple forms of tumour antigen and its loading, a wide range of adjuvants and different modes of vaccine delivery. Key insights from pre-clinical studies, and lessons learned from early clinical testing drive progress towards improved vaccines. The potential to fortify responses with other modalities of immunotherapy makes clinically effective "second generation" DC vaccination strategies a priority for cancer immune therapists.
Collapse
Affiliation(s)
- Christian E Bryant
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia.
| | - Sarah Sutherland
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Benjamin Kong
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Michael S Papadimitrious
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Derek N J Hart
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia.
| |
Collapse
|
224
|
Immune signatures predicting responses to immunomodulatory antibody therapy. Curr Opin Immunol 2018; 51:91-96. [PMID: 29550661 DOI: 10.1016/j.coi.2018.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 02/02/2018] [Accepted: 03/01/2018] [Indexed: 12/30/2022]
Abstract
Since the first immunomodulatory antibody was licensed by the FDA in 2011 for treating melanoma it has remained the case that only a certain proportion of cancer patients respond favourably to a particular therapy. Recent results from combining two or more different antibodies each targeting a different immune checkpoint indicate that the proportion of responding patients can be increased, but thus far there are no such therapies routinely yielding clinical benefit in 100% of patients in any cancer type. Therefore, predicting which patients will respond to a particular therapy remains of the utmost importance in order to maximise treatment efficacy and minimise side-effects and costs. Moreover, determining biomarkers predicting responses may provide insight into the mechanisms responsible for success or failure of that therapy. This article reviews seminal papers mostly from the past two years of progress in this area of intense investigation, and mostly in melanoma, the tumour type for which the largest body of data exists thus far.
Collapse
|
225
|
Anti-programmed cell death protein 1 tolerance and efficacy after ipilimumab immunotherapy: observational study of 39 patients. Melanoma Res 2018; 27:110-115. [PMID: 27926587 DOI: 10.1097/cmr.0000000000000313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In patients with ipilimumab (IPI)-refractory melanoma, the anti-programmed cell death proteins 1 (PD1s) nivolumab (NIV) and pembrolizumab (PEM) are considered to be a new standard of treatment. Few data are available on anti-PD1 safety in patients who develop IPI-related severe adverse events (AEs) (grade≥3). The aim of this study was to compare the anti-PD1 safety and efficacy in patients with previous severe toxicity to IPI versus in those showing moderate and no previous IPI-related AEs. This single institution-based observational study included all patients treated with anti-PD1 (PEM or NIV) and previously treated with IPI for unresectable stage III or IV melanoma. The patients enrolled were classified according to the occurrence of IPI-related AEs: group A: no previous IPI-related AEs; group B: mild to moderate IPI-related AEs; and group C: severe to life-threatening IPI-related AEs. The main outcome measure was safety of the anti-PD1 among the three groups. The secondary endpoints included response parameters. Groups A, B, and C included, respectively, 16, 13, and 10 patients. The incidence of severe anti-PD1-related AEs (grades 3-4) was 12, 23, and 10% in groups A, B, and C, respectively. One-year estimates of survival were 52.2, 73.4, and 66.7% among the patients in groups A, B, and C, respectively. The number of patients was too small to enable a meaningful statistical comparison. We did not observe any difference in anti-PD1 toxicity onset incidence according to the occurrence of previous IPI AEs. These reassuring real-life data should be confirmed in a wider analysis.
Collapse
|
226
|
Bello DM, Ariyan CE. Adjuvant Therapy in the Treatment of Melanoma. Ann Surg Oncol 2018; 25:1807-1813. [PMID: 29468608 DOI: 10.1245/s10434-018-6376-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Danielle M Bello
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Charlotte E Ariyan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
227
|
Gonzalez-Cao M, Boada A, Teixidó C, Fernandez-Figueras MT, Mayo C, Tresserra F, Bustamante J, Viteri S, Puertas E, Santarpia M, Riso A, Barron F, Karachaliou N, Rosell R. Fatal gastrointestinal toxicity with ipilimumab after BRAF/MEK inhibitor combination in a melanoma patient achieving pathological complete response. Oncotarget 2018; 7:56619-56627. [PMID: 27447748 PMCID: PMC5302939 DOI: 10.18632/oncotarget.10651] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 05/29/2016] [Indexed: 01/08/2023] Open
Abstract
Approximately 50% of metastatic melanoma patients harbor BRAF mutations. Several treatment options including the combination of BRAF and MEK inhibitors (BRAF/MEKi) and immunotherapy (mainly anti CTLA-4 and anti PD-1 antibodies), have been shown to improve survival in these patients. Although preclinical data support the synergistic effect of both modalities in combination, data confirming the activity and tolerability of these combinations are not yet available in the clinical setting. Herein, we report the case of a melanoma patient treated with sequential BRAF/MEKi (dabrafenib plus trametinib) followed by the anti CTLA-4 antibody ipilimumab who achieved a pathological complete response. Unfortunately, the patient died due to fatal gastrointestinal (GI) toxicity. Analysis of the BRAFV600E mutation in circulating tumoral DNA (ctDNA) from peripheral blood samples and serial tumor tissue biopsies throughout treatment demonstrated a good correlation with clinical evolution.
Collapse
Affiliation(s)
- Maria Gonzalez-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain
| | - Aram Boada
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Cristina Teixidó
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain.,Pangaea Biotech, Laboratory of Oncology, Barcelona, Spain
| | | | - Clara Mayo
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain.,Pangaea Biotech, Laboratory of Oncology, Barcelona, Spain
| | - Francesc Tresserra
- Pathology Department, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain
| | | | - Santiago Viteri
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain
| | - Enrique Puertas
- Radiotherapy Department, Hospital Quirónsalud, Barcelona, Spain
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy
| | - Aldo Riso
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain
| | - Feliciano Barron
- Medical Oncology Unit, Insituto Nacional de Cancerología, México
| | - Niki Karachaliou
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain
| | - Rafael Rosell
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital-Quirónsalud Group, Barcelona, Spain.,Catalan Institute of Oncology, Cancer Biology & Precision Medicine Programme, Germans Trias i Pujol Hospital and Health Sciences Institute, Badalona, Spain
| |
Collapse
|
228
|
Frosch ZAK, Gagne JJ, Gray SW, Abel GA. What Does a Cancer Diagnosis Mean? Public Expectations in a Shifting Therapeutic Environment. J Oncol Pract 2018; 14:139-140. [PMID: 29381410 DOI: 10.1200/jop.17.00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Zachary A K Frosch
- Dana-Farber Cancer Institute; Brigham and Women's Hospital, Boston, MA; and City of Hope, Duarte, CA
| | - Joshua J Gagne
- Dana-Farber Cancer Institute; Brigham and Women's Hospital, Boston, MA; and City of Hope, Duarte, CA
| | - Stacy W Gray
- Dana-Farber Cancer Institute; Brigham and Women's Hospital, Boston, MA; and City of Hope, Duarte, CA
| | - Gregory A Abel
- Dana-Farber Cancer Institute; Brigham and Women's Hospital, Boston, MA; and City of Hope, Duarte, CA
| |
Collapse
|
229
|
Shreders A, Joseph RW. Efficacy of immunotherapy for metastatic mucosal melanoma. Immunotherapy 2018; 8:843-5. [PMID: 27381682 DOI: 10.2217/imt-2016-0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
230
|
Belzile O, Huang X, Gong J, Carlson J, Schroit AJ, Brekken RA, Freimark BD. Antibody targeting of phosphatidylserine for the detection and immunotherapy of cancer. Immunotargets Ther 2018; 7:1-14. [PMID: 29417044 PMCID: PMC5788995 DOI: 10.2147/itt.s134834] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Phosphatidylserine (PS) is a negatively charged phospholipid in all eukaryotic cells that is actively sequestered to the inner leaflet of the cell membrane. Exposure of PS on apoptotic cells is a normal physiological process that triggers their rapid removal by phagocytic engulfment under noninflammatory conditions via receptors primarily expressed on immune cells. PS is aberrantly exposed in the tumor microenvironment and contributes to the overall immunosuppressive signals that antagonize the development of local and systemic antitumor immune responses. PS-mediated immunosuppression in the tumor microenvironment is further exacerbated by chemotherapy and radiation treatments that result in increased levels of PS on dying cells and necrotic tissue. Antibodies targeting PS localize to tumors and block PS-mediated immunosuppression. Targeting exposed PS in the tumor microenvironment may be a novel approach to enhance immune responses to cancer.
Collapse
Affiliation(s)
- Olivier Belzile
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Xianming Huang
- Department of Preclinical Research.,Department of Antibody Discovery, Peregrine Pharmaceuticals, Inc., Tustin, CA, USA
| | - Jian Gong
- Department of Preclinical Research.,Department of Antibody Discovery, Peregrine Pharmaceuticals, Inc., Tustin, CA, USA
| | - Jay Carlson
- Department of Preclinical Research.,Department of Antibody Discovery, Peregrine Pharmaceuticals, Inc., Tustin, CA, USA
| | - Alan J Schroit
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bruce D Freimark
- Department of Preclinical Research.,Department of Antibody Discovery, Peregrine Pharmaceuticals, Inc., Tustin, CA, USA
| |
Collapse
|
231
|
Melanoma: Immunotherapy in Advanced Melanoma and in the Adjuvant Setting. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
232
|
Iglesias P. Cancer immunotherapy-induced endocrinopathies: Clinical behavior and therapeutic approach. Eur J Intern Med 2018; 47:6-13. [PMID: 28826822 DOI: 10.1016/j.ejim.2017.08.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022]
Abstract
Cancer immunotherapy has proven to be effective in a wide variety of tumors. The use of immune checkpoint blocking monoclonal antibodies has become a standard treatment regimen in some of them as advanced melanoma. However, given the mechanism of action, its use may be associated with immune-related adverse events that may complicate the clinical course and prognosis of patients. Among these are autoimmune endocrine adverse effects, such as hypophysitis, hypo and hyperthyroidism, and adrenal insufficiency. This review focuses on the most relevant and new aspects related to the incidence, clinical presentation, diagnosis and treatment of these adverse effects associated with different types of immune checkpoint inhibitors in cancer immunotherapy.
Collapse
Affiliation(s)
- Pedro Iglesias
- Department of Endocrinology, Hospital Ramón y Cajal, Ctra. de Colmenar, Km 9.100, 28034 Madrid, Spain.
| |
Collapse
|
233
|
Borrie AE, Maleki Vareki S. T Lymphocyte–Based Cancer Immunotherapeutics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:201-276. [DOI: 10.1016/bs.ircmb.2018.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
234
|
Veenstra R, Kostine M, Cleton-Jansen AM, de Miranda NF, Bovée JV. Immune checkpoint inhibitors in sarcomas: in quest of predictive biomarkers. J Transl Med 2018; 98:41-50. [PMID: 29155424 DOI: 10.1038/labinvest.2017.128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/21/2017] [Accepted: 09/24/2017] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are a rare group of tumors of mesenchymal origin. Metastatic sarcomas are often difficult to treat and unresponsive to standard radio- and chemotherapy, resulting in a poor survival rate for patients. Novel treatments with immune checkpoint inhibitors have been proven to prolong survival of patients with a variety of cancers, including metastatic melanoma, lung, and renal cell carcinoma. Since immune checkpoint inhibitors could provide a novel treatment option for patients with sarcomas, clinical trials investigating their efficacy in these group of tumors are ongoing. However, the discrimination of patients that are the most likely to respond to these treatments is still an obstacle in the design of clinical trials. In this review, we provide a brief overview of the mechanisms of action of immune checkpoint inhibitors and discuss the proposed biomarkers of therapy response, such as lymphocytic infiltration, intratumoral PD-L1 expression, and mutational load in sarcomas.
Collapse
Affiliation(s)
- Robin Veenstra
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Kostine
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Noel Fcc de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Judith Vmg Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
235
|
Calabrò L, Rossi G, Maio M. New horizons from immunotherapy in malignant pleural mesothelioma. J Thorac Dis 2018; 10:S322-S332. [PMID: 29507802 PMCID: PMC5830566 DOI: 10.21037/jtd.2017.12.88] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive disease with a severe prognosis. Medical treatment for MPM unresectable patients is still unsatisfactory; therefore novel therapeutic approaches are urgently needed. Immunotherapy represents a promising treatment for MPM patients. Here, we'll discuss the most promising immunotherapeutic treatments currently under active investigation for this still dreadful disease.
Collapse
Affiliation(s)
- Luana Calabrò
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Giulia Rossi
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| |
Collapse
|
236
|
State-of-the-Art Diagnosis and Treatment of Melanoma. J Comput Assist Tomogr 2018; 42:331-339. [DOI: 10.1097/rct.0000000000000697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
237
|
Elliott TM, Whiteman DC, Olsen CM, Gordon LG. Estimated Healthcare Costs of Melanoma in Australia Over 3 Years Post-Diagnosis. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2017; 15:805-816. [PMID: 28756584 DOI: 10.1007/s40258-017-0341-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
BACKGROUND Skin cancer exerts a large and growing burden on health systems. With new pharmacotherapies for metastatic melanoma now available, a contemporary understanding of the cost burden of melanoma control is warranted. OBJECTIVE To comprehensively assess the healthcare costs of malignant melanoma diagnosis and treatment in Australia, over 3 years after diagnosis. METHODS We developed a decision-analytic model and micro-costing method to estimate the mean cost per patient for melanoma, incorporating all diagnostic and treatment modalities used in Australia (2017 AU$). By using the de-identified 10% sample of Medicare Benefits Scheme, we analysed health service use and supplemented our analyses with published estimates. We took a health system cost perspective, and addressed input uncertainty with sensitivity analyses. RESULTS The mean annual cost per patient for melanoma stage 0/I/II was AU$1681 (US$1175) rising to AU$37,729 (US$26,365) for stage III resectable, and AU$115,109 (US$80,440) for stage III unresectable/IV. Three-year costs for stage III unresectable/IV were AU$187,720. Nationally, the annual estimated cost for treatment of all new cases of in situ and invasive melanomas was AU$201 million (95% CI: AU$187 to AU$216 million). When we included treatments for presumptive melanoma later found to be benign lesions, the estimated annual cost burden reached AU$272 million. CONCLUSION With rapidly rising treatment costs, there is a need to consider a comprehensive melanoma control strategy that includes primary prevention of skin cancers and cost-effective sun protection initiatives.
Collapse
Affiliation(s)
- Thomas M Elliott
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Q4006, Australia
- Menzies Health Institute Queensland, Centre for Applied Health Economics, Griffith University, Logan Campus, University Dr, Meadowbrook, Q4131, Australia
- Centre for Research Excellence in Sun and Health Queensland, University of Technology, Victoria Park Rd, Kelvin Grove, Brisbane, Q4059, Australia
| | - David C Whiteman
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Q4006, Australia
- Centre for Research Excellence in Sun and Health Queensland, University of Technology, Victoria Park Rd, Kelvin Grove, Brisbane, Q4059, Australia
| | - Catherine M Olsen
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Q4006, Australia
- School of Public Health, The University of Queensland, Brisbane, Australia
| | - Louisa G Gordon
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Q4006, Australia.
- Centre for Research Excellence in Sun and Health Queensland, University of Technology, Victoria Park Rd, Kelvin Grove, Brisbane, Q4059, Australia.
- School of Public Health, The University of Queensland, Brisbane, Australia.
- School of Nursing, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
238
|
Carter PJ, Lazar GA. Next generation antibody drugs: pursuit of the 'high-hanging fruit'. Nat Rev Drug Discov 2017; 17:197-223. [DOI: 10.1038/nrd.2017.227] [Citation(s) in RCA: 559] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
239
|
Affiliation(s)
- Vinay Prasad
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland
| |
Collapse
|
240
|
Amaral T, Meraz-Torres F, Garbe C. Immunotherapy in managing metastatic melanoma: which treatment when? Expert Opin Biol Ther 2017; 17:1523-1538. [PMID: 28891339 DOI: 10.1080/14712598.2017.1378640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Ten to fifteen percent of melanoma patients develop distant or unresectable metastasis requiring systemic treatment. Around 45% of the patients diagnosed with metastatic cutaneous melanoma harbor a BRAFV600 mutation and derive benefit from combined targeted therapy with MAPK pathway inhibitors. These offer a rapid response that translates into improvement of symptoms and increased quality of life. However, resistance often develops with subsequent progressive disease. Immunotherapy with checkpoint inhibitors may be offered to BRAF-mutated and wild-type patients and is associated with longer and durable responses that can continue over years. Areas covered: In this review, the authors discuss the late evidence for targeted and immunotherapy in melanoma patients, as well as therapy sequencing. Immunotherapy in special populations is also addressed. Expert opinion: Effective treatments are currently available. However, there are still unanswered questions of the best therapy sequence, the clear superiority of combined immunotherapy versus monotherapy in all patients, and therapy duration. Since different promising treatments will become available, clinical trials comparing the diverse options in terms of safety, efficacy and cost- effectiveness are required to make the right decisions. Consequently, patients should be encouraged to participate in clinical trials, whenever possible.
Collapse
Affiliation(s)
- Teresa Amaral
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
- b Portuguese Air Force Health Direction , Lisbon , Portugal
| | - Francisco Meraz-Torres
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
| | - Claus Garbe
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
| |
Collapse
|
241
|
Jamal R, Lapointe R, Cocolakis E, Thébault P, Kazemi S, Friedmann JE, Dionne J, Cailhier JF, Bélanger K, Ayoub JP, Le H, Lambert C, El-Hajjar J, van Kempen LC, Spatz A, Miller WH. Peripheral and local predictive immune signatures identified in a phase II trial of ipilimumab with carboplatin/paclitaxel in unresectable stage III or stage IV melanoma. J Immunother Cancer 2017; 5:83. [PMID: 29157311 PMCID: PMC5696743 DOI: 10.1186/s40425-017-0290-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Checkpoint blockade with ipilimumab provides long-term survival to a significant proportion of patients with metastatic melanoma. New approaches to increase survival and to predict which patients will benefit from treatment are needed. This phase II trial combined ipilimumab with carboplatin/paclitaxel (CP) to assess its safety, efficacy, and to search for peripheral and tumor-based predictive biomarkers. METHODS Thirty patients with untreated unresectable/metastatic melanoma were treated with ipilimumab and CP. Adverse events (AEs) were monitored and response to treatment was evaluated. Tumor tissue and peripheral blood were collected at specified time points to characterize tumor immune markers by immunohistochemistry and systemic immune activity by multiplex assays and flow cytometry. RESULTS Eighty three percent of patients received all 5 cycles of CP and 93% completed ipilimumab induction. Serious AEs occurred in 13% of patients, and no treatment-related deaths were observed. Best Overall Response Rate (BORR) and Disease Control Rate (DCR) were 27 and 57%, respectively. Median overall survival was 16.2 months. Response to treatment was positively correlated with a higher tumor CD3+ infiltrate (immune score) at baseline. NRAS and BRAF mutations were less frequent in patients who experienced clinical benefit. Assessment of peripheral blood revealed that non-responders had elevated baseline levels of CXCL8 and CCL4, and a higher proportion of circulating late differentiated B cells. Pre-existing high levels of chemokines (CCL3, CCL4 and CXCL8) and advanced B cell differentiation were strongly associated with worse patient overall survival. Elevated proportions of circulating CD8+/PD-1+ T cells during treatment were associated with worse survival. CONCLUSIONS The combination of ipilimumab and CP was well tolerated and revealed novel characteristics associated with patients likely to benefit from treatment. A pre-existing systemic inflammatory state characterized by elevation of selected chemokines and advanced B cell differentiation, was strongly associated with poor patient outcomes, revealing potential predictive circulating biomarkers. TRIAL REGISTRATION Clinicaltrials.gov , NCT01676649 , registered on August 29, 2012.
Collapse
Affiliation(s)
- Rahima Jamal
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Réjean Lapointe
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Eftihia Cocolakis
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Paméla Thébault
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Shirin Kazemi
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jennifer E. Friedmann
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jeanne Dionne
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Jean-François Cailhier
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Karl Bélanger
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Jean-Pierre Ayoub
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Huy Le
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Caroline Lambert
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jida El-Hajjar
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Léon C. van Kempen
- Department of Pathology, Molecular Pathology Center, Jewish General Hospital, McGill University, Montreal, QC Canada
| | - Alan Spatz
- Department of Pathology, Molecular Pathology Center, Jewish General Hospital, McGill University, Montreal, QC Canada
| | - Wilson H. Miller
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| |
Collapse
|
242
|
Marra A, Scognamiglio G, Peluso I, Botti G, Fusciello C, Filippelli A, Ascierto PA, Pepe S, Sabbatino F. Immune Checkpoint Inhibitors in Melanoma and HIV Infection. Open AIDS J 2017; 11:91-100. [PMID: 29290886 PMCID: PMC5730951 DOI: 10.2174/1874613601711010091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022] Open
Abstract
Introduction: Immunotherapy with immune checkpoint inhibitors increases the overall survival of patients with metastatic melanoma regardless of their oncogene addicted mutations. However, no data is available from clinical trials of effective therapies in subgroups of melanoma patients that carry chronic infective diseases such as HIV. Evidences suggest a key role of the immune checkpoint molecules as a mechanism of immune escape not only from melanoma but also from HIV host immune response. Conclusion: In this article, firstly, we will describe the role of the immune checkpoint molecules in HIV chronic infection. Secondly, we will summarize the most relevant clinical evidences utilizing immune checkpoint inhibitors for the treatment of melanoma patients. Lastly, we will discuss the potential implications as well as the potential applications of immune checkpoint molecule-based immunotherapy in patients with melanoma and HIV infection.
Collapse
Affiliation(s)
- Antonio Marra
- Department of Medical Oncology, San Gerardo Hospital, via G. B. Pergolesi, 20052 Monza, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Ilaria Peluso
- Hematology Unit, Department of Clinical and Surgical Medicine, University of Naples Federico II, via S. Pansini, 80131 Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Celeste Fusciello
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Amelia Filippelli
- Pharmacology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Stefano Pepe
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| |
Collapse
|
243
|
|
244
|
Kukar M, Gabriel E, May R, Cho E, Lichtenthal M, Groman A, Skitzki J, Francescutti V, Kane JM. Conditional Survival-Based “Abbreviated” Routine Cancer Surveillance for Pathologic Stage IB Melanoma. Am Surg 2017. [DOI: 10.1177/000313481708301128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A negative sentinel lymph node biopsy (SLNB) for stage IB (T1b/T2a N0) melanoma would predict an excellent long-term prognosis. Combined with the concept of conditional survival, an “abbreviated” cancer surveillance strategy was implemented to reduce the number of visits and total length of follow-up. Retrospective review of all pathologic stage IB melanoma patients (negative SLNB) at a single institution between 2006 and 2008 after implementation of an “abbreviated” cancer surveillance; clinic visits every six months for five years followed by one annual visit (total follow-up six years). Patient demographics, tumor characteristics, and information regarding recurrences were obtained. Recurrence-free, disease-specific, and overall survival were calculated. Eighty-seven patients underwent the “abbreviated” cancer surveillance. Median age was 55.4 years and 50.6 per cent were male. Median Breslow thickness was 1.1 mm (range 0.5–2.0 mm) and 1.1 per cent were ulcerated. Primary tumor site was 49 per cent extremities, 39 per cent trunk, and 12 per cent head/ neck. Median follow-up was 68.6 months. Five-year recurrence-free, disease-specific, and overall survivals were 89, 95, and 88 per cent, respectively. During surveillance, 10 patients had concerning symptoms or physical findings prompting subsequent workup, all of which were negative for recurrence/metastases. There were only three true melanoma recurrences; all were distant metastases and presented symptomatically between scheduled follow-up visits. In light of the excellent prognosis for pathologic (SLNB negative) stage IB melanoma, an “abbreviated” cancer surveillance schedule based on conditional survival would reduce both direct and indirect costs in this cohort. The few recurrences were symptomatic and unlikely to have changed with more intensive surveillance.
Collapse
Affiliation(s)
- Moshim Kukar
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Emmanuel Gabriel
- Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, Jacksonville, Florida; and the
| | - Rebecca May
- Department of Surgery, State University of New York, Buffalo, New York
| | - Edward Cho
- Department of Surgery, State University of New York, Buffalo, New York
| | - Michelle Lichtenthal
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Adrienne Groman
- Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, New York
| | - Joseph Skitzki
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Valerie Francescutti
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - John M. Kane
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
245
|
Adoptive cell therapy with CD4 + T helper 1 cells and CD8 + cytotoxic T cells enhances complete rejection of an established tumour, leading to generation of endogenous memory responses to non-targeted tumour epitopes. Clin Transl Immunology 2017; 6:e160. [PMID: 29114389 PMCID: PMC5671987 DOI: 10.1038/cti.2017.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 01/31/2023] Open
Abstract
The results of adoptive T-cell therapies (ACTs) are very encouraging and show clinical evidence that ACT can provide a cure for patients with metastatic disease. However, various response rates and long-term cancer remission have been observed in different ACT trials. The types of T cells, prior treatment with chemotherapy and co-administration of other immune-target therapies have been found to influence the efficacy of ACT. In this study, we investigate the ability of ACT using CD4+ T helper 1 (Th1) cells and CD8+ cytotoxic T lymphocytes (CTLs) to reject the growth of established B16-ovalbumin (OVA) melanoma. CD8+ CTLs were found to be the main effector T cells that mediated tumour regression. However, low tumour-free survival rates were observed in ACT with CD8+ CTLs only. Co-transferring CD4+ Th1 cells and CD8+ CTLs has been observed to induce a synergistic antitumour response, resulting in complete regression in 80% of the tumour-bearing mice. We also examined a prior Dacarbazine (DTIC) and after virus-like particle (VLP)-OVA vaccine treatment to enhance ACT, but no therapeutic benefit was observed during primary B16-OVA tumour growth. Nevertheless, the ACT-mediated antitumour response was able to generate memory responses to both B16-OVA and B16-gp33 tumours. VLP-OVA vaccination following ACT enhances the memory responses to tumours that express a heterogenic population of both B16-OVA and B16-gp33 cells; however, it abolished the memory response to tumours consisting of only gp33-expressing cells. These findings provide important information for designing therapeutic treatments for patients with metastatic disease and cancer relapse to achieve durable cancer remission.
Collapse
|
246
|
Checkpoint Inhibitor-Induced Colitis: A New Type of Inflammatory Bowel Disease? ACG Case Rep J 2017; 4:e112. [PMID: 29043290 PMCID: PMC5636906 DOI: 10.14309/crj.2017.112] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Checkpoint inhibitors are immune-stimulatory antibodies that have transformed the management and prognosis of individuals with metastatic melanoma and other cancers. Checkpoint inhibitor–induced colitis is an increasingly recognized immune-related adverse event that shares many of the same phenotypical, serological, and histological characteristics of both Crohn’s disease and ulcerative colitis, suggesting that checkpoint inhibitor–induced colitis may represent a new inflammatory bowel disease phenotype. We report a 73-year-old man with metastatic melanoma who developed ipilimumab-induced colitis with subsequent transformation to Crohn?s colitis-like phenotype after the addition of pembrolizumab.
Collapse
|
247
|
Kranz LM, Beck JD, Grunwitz C, Hotz C, Vormehr M, Diken M. CIMT 2017: Anniversary symposium - Report on the 15th CIMT Annual Meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2017; 13:2272-2279. [PMID: 28846471 PMCID: PMC5647989 DOI: 10.1080/21645515.2017.1358327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Lena M Kranz
- a BioNTech RNA Pharmaceuticals GmbH , Mainz , Germany
| | - Jan D Beck
- b TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| | | | | | - Mathias Vormehr
- c BioNTech AG , Mainz , Germany.,d Research Center for Immunotherapy (FZI), University Medical Center at the Johannes Gutenberg University Mainz , Mainz , Germany
| | - Mustafa Diken
- b TRON-Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH , Mainz , Germany
| |
Collapse
|
248
|
Advances in the Immunobiological Therapies for Advanced Melanoma. ACTAS DERMO-SIFILIOGRAFICAS 2017. [DOI: 10.1016/j.adengl.2017.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
249
|
Rangel-Sosa MM, Aguilar-Córdova E, Rojas-Martínez A. Immunotherapy and gene therapy as novel treatments for cancer. COLOMBIA MEDICA (CALI, COLOMBIA) 2017; 48:138-147. [PMID: 29213157 PMCID: PMC5687866 DOI: 10.25100/cm.v48i3.2997] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The immune system interacts closely with tumors during the disease development and progression to metastasis. The complex communication between the immune system and the tumor cells can prevent or promote tumor growth. New therapeutic approaches harnessing protective immunological mechanisms have recently shown very promising results. This is performed by blocking inhibitory signals or by activating immunological effector cells directly. Immune checkpoint blockade with monoclonal antibodies directed against the inhibitory immune receptors CTLA-4 and PD-1 has emerged as a successful treatment approach for patients with advanced melanoma. Ipilimumab is an anti-CTLA-4 antibody which demonstrated good results when administered to patients with melanoma. Gene therapy has also shown promising results in clinical trials. Particularly, Herpes simplex virus (HSV)-mediated delivery of the HSV thymidine kinase (TK) gene to tumor cells in combination with ganciclovir (GCV) may provide an effective suicide gene therapy for destruction of glioblastomas, prostate tumors and other neoplasias by recruiting tumor-infiltrating lymphocytes into the tumor. The development of new treatment strategies or combination of available innovative therapies to improve cell cytotoxic T lymphocytes trafficking into the tumor mass and the production of inhibitory molecules blocking tumor tissue immune-tolerance are crucial to improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Martha Montserrat Rangel-Sosa
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León. Nuevo León, México
| | | | | |
Collapse
|
250
|
Dinnes J, Saleh D, Newton-Bishop J, Cheung ST, Nathan P, Matin RN, Chuchu N, Bayliss SE, Takwoingi Y, Davenport C, Godfrey K, O'Sullivan C, Deeks JJ, Williams HC. Tests to assist in the staging of cutaneous melanoma: a generic protocol. Hippokratia 2017. [DOI: 10.1002/14651858.cd012806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Jacqueline Dinnes
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Daniel Saleh
- Newcastle Hospitals NHS Trust, Royal Victoria Infirmary; Newcastle Hospitals; Newcastle UK
| | - Julia Newton-Bishop
- University of Leeds; Section of Epidemiology and Biostatistics; St James's Hospital Leeds UK LS9 7TF
| | - Seau Tak Cheung
- Dudley Hospitals Foundation Trust, Corbett Hospital; Department of Dermatology; Wicarage Road Stourbridge UK DY8 4JB
| | - Paul Nathan
- Mount Vernon Hospital; Mount Vernon Cancer Centre; Rickmansworth Road Northwood UK HA6 2RN
| | - Rubeta N Matin
- Churchill Hospital; Department of Dermatology; Old Road Headington Oxford UK OX3 7LJ
| | - Naomi Chuchu
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Susan E Bayliss
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Yemisi Takwoingi
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Clare Davenport
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Kathie Godfrey
- The University of Nottingham; c/o Cochrane Skin Group; Nottingham UK
| | | | - Jonathan J Deeks
- University of Birmingham; Institute of Applied Health Research; Birmingham UK B15 2TT
| | - Hywel C Williams
- The University of Nottingham; Centre of Evidence Based Dermatology; Queen's Medical Centre Derby Road Nottingham UK NG7 2UH
| |
Collapse
|