101
|
Huang YF, Xie WJ, Fan HY, Du J. Comparative Risks of High-Grade Adverse Events Among FDA-Approved Systemic Therapies in Advanced Melanoma: Systematic Review and Network Meta-Analysis. Front Oncol 2020; 10:571135. [PMID: 33178599 PMCID: PMC7593404 DOI: 10.3389/fonc.2020.571135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/07/2020] [Indexed: 01/03/2023] Open
Abstract
Background: Head-to-head evidence is lacking in comparative risks of high-grade adverse events (AEs) among different systemic treatment options for advanced melanoma. Methods: An up-to-date systematic review and network meta-analysis (NMA) was performed. Randomized controlled trials (RCTs) of patients with advanced melanoma were eligible if at least one intervention was the Food and Drug Administration-approved targeted or immune checkpoint inhibitors. Risks of high-grade AEs were estimated by random-effects Bayesian NMAs, based on relative risks. Surface under the cumulative ranking probabilities was used to assess relative ranking of treatments. The summary incidences were calculated. Results: Twenty-five RCTs (12,925 patients) comparing 10 different systemic treatment options were included. BRAF/MEK had the highest risk of overall high-grade AEs (pooled incidence: 32.11%). BRAF had the highest risk of high-grade arthralgia (0.39%), whereas MEK had the highest risk of high-grade hypertension (2.28%) and nausea (0.37%). Cytotoxic T-lymphocyte antigen 4 (CTLA-4)/chemo had the highest risk of high-grade diarrhea (1.31%), alanine aminotransferase (0.60%), and aspartate aminotransferase elevation (0.59%). Programmed cell death 1 (PD-1)/CTLA-4 had the highest risks of high-grade pyrexia (1.14%) and rash (0.94%). Using PD-1 inhibitor alone had the lowest risks of overall high-grade AEs. Conclusions: Different systemic treatment options have varying high-grade AEs in advanced melanoma treatment. Current evidences highlight the important risks of BRAF/MEK, CTLA-4/chemo, and PD-1/CTLA-4.
Collapse
Affiliation(s)
- Ya-Fang Huang
- School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| | - Wen-Jie Xie
- Department Clinical Research, University of Bern, Bern, Switzerland
| | - Hai-Yu Fan
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Juan Du
- School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| |
Collapse
|
102
|
Saleh R, Toor SM, Elkord E. Targeting TIM-3 in solid tumors: innovations in the preclinical and translational realm and therapeutic potential. Expert Opin Ther Targets 2020; 24:1251-1262. [PMID: 33103506 DOI: 10.1080/14728222.2020.1841750] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have shown a great therapeutic efficacy in cancer patients. However, a significant proportion of cancer patients remain unresponsive or show limited response. T cell immunoglobulin and mucin-domain containing protein-3 (TIM-3) is a co-inhibitory receptor expressed on various cell types and is involved in the attenuation of immune responses. TIM-3 and its ligands are highly expressed in various solid malignancies and some studies have reported its association with worse disease outcomes. Thus, targeting TIM-3 could be a promising therapeutic approach to treat cancer patients. AREAS COVERED This review describes the role of TIM-3 and its ligands in regulating anti-tumor immunity and their contribution to cancer progression. Moreover, this review focuses on the preclinical models and translational data from important studies published in PubMed till October 2020, which demonstrate the therapeutic benefits of targeting TIM-3 signaling. EXPERT OPINION Despite the promising data obtained from targeting TIM-3 in preclinical models, precise mechanisms underlying the anti-tumor effects of TIM-3 inhibition are not fully elucidated. Therefore, mechanistic studies are required to provide better insights into the anti-tumor effects of targeting TIM-3, and clinical data are necessary to determine the safety profiles and therapeutic efficacy of TIM-3 inhibition in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford , Manchester, United Kingdom
| |
Collapse
|
103
|
Tran KB, Buchanan CM, Shepherd PR. Evolution of Molecular Targets in Melanoma Treatment. Curr Pharm Des 2020; 26:396-414. [PMID: 32000640 DOI: 10.2174/1381612826666200130091318] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
Melanoma is the deadliest type of skin cancers, accounting for more than 80% of skin cancer mortality. Although melanoma was known very early in the history of medicine, treatment for this disease had remained largely the same until very recently. Previous treatment options, including removal surgery and systemic chemotherapy, offered little benefit in extending the survival of melanoma patients. However, the last decade has seen breakthroughs in melanoma treatment, which all emerged following new insight into the oncogenic signaling of melanoma. This paper reviewed the evolution of drug targets for melanoma treatment based on the emergence of novel findings in the molecular signaling of melanoma. One of the findings that are most influential in melanoma treatment is that more than 50% of melanoma tumors contain BRAF mutations. This is fundamental for the development of BRAF inhibitors, which is the first group of drugs that significantly improves the overall survival of melanoma patients compared to the traditional chemotherapeutic dacarbazine. More recently, findings of the role of immune checkpoint molecules such as CTLA-4 and PD1/PD-L1 in melanoma biology have led to the development of a new therapeutic category: immune checkpoint inhibitors, which, for the first time in the history of cancer treatment, produced a durable response in a subset of melanoma patients. However, as this paper discussed next, there is still an unmet need for melanoma treatment. A significant population of patients did not respond to either BRAF inhibitors or immune checkpoint inhibitors. Of those patients who gained an initial response from those therapies, a remarkable percentage would develop drug resistance even when MEK inhibitors were added to the treatment. Finally, this paper discusses some possible targets for melanoma treatment.
Collapse
Affiliation(s)
- Khanh B Tran
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Christina M Buchanan
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| |
Collapse
|
104
|
Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020; 20:651-668. [PMID: 32433532 PMCID: PMC7238960 DOI: 10.1038/s41577-020-0306-5] [Citation(s) in RCA: 2572] [Impact Index Per Article: 514.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
The T lymphocyte, especially its capacity for antigen-directed cytotoxicity, has become a central focus for engaging the immune system in the fight against cancer. Basic science discoveries elucidating the molecular and cellular biology of the T cell have led to new strategies in this fight, including checkpoint blockade, adoptive cellular therapy and cancer vaccinology. This area of immunological research has been highly active for the past 50 years and is now enjoying unprecedented bench-to-bedside clinical success. Here, we provide a comprehensive historical and biological perspective regarding the advent and clinical implementation of cancer immunotherapeutics, with an emphasis on the fundamental importance of T lymphocyte regulation. We highlight clinical trials that demonstrate therapeutic efficacy and toxicities associated with each class of drug. Finally, we summarize emerging therapies and emphasize the yet to be elucidated questions and future promise within the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Alex D Waldman
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jill M Fritz
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
105
|
Kuryk L, Bertinato L, Staniszewska M, Pancer K, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers (Basel) 2020; 12:cancers12103057. [PMID: 33092131 PMCID: PMC7589099 DOI: 10.3390/cancers12103057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Here, we review the current state of knowledge in the field of cancer immunotherapy, focusing on the scientific rationale for the use of oncolytic viruses, checkpoint inhibitors and their combination to combat melanomas. Attention is also given to the immunological aspects of cancer therapy and the shift from conventional therapy towards immunotherapy. This review brings together information on how immunotherapy can be applied to support other cancer therapies in order to maximize the efficacy of melanoma treatment and improve clinical outcomes. Abstract In this review, we discuss the use of oncolytic viruses and checkpoint inhibitors in cancer immunotherapy in melanoma, with a particular focus on combinatory therapies. Oncolytic viruses are promising and novel anti-cancer agents, currently under investigation in many clinical trials both as monotherapy and in combination with other therapeutics. They have shown the ability to exhibit synergistic anticancer activity with checkpoint inhibitors, chemotherapy, radiotherapy. A coupling between oncolytic viruses and checkpoint inhibitors is a well-accepted strategy for future cancer therapies. However, eradicating advanced cancers and tailoring the immune response for complete tumor clearance is an ongoing problem. Despite current advances in cancer research, monotherapy has shown limited efficacy against solid tumors. Therefore, current improvements in virus targeting, genetic modification, enhanced immunogenicity, improved oncolytic properties and combination strategies have a potential to widen the applications of immuno-oncology (IO) in cancer treatment. Here, we summarize the strategy of combinatory therapy with an oncolytic vector to combat melanoma and highlight the need to optimize current practices and improve clinical outcomes.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
- Clinical Science, Targovax Oy, Saukonpaadenranta 2, 00180 Helsinki, Finland
- Correspondence: (L.K.); (M.G.)
| | - Laura Bertinato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Monika Staniszewska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
- Correspondence: (L.K.); (M.G.)
| |
Collapse
|
106
|
Xu Y, Fu Y, Zhu B, Wang J, Zhang B. Predictive Biomarkers of Immune Checkpoint Inhibitors-Related Toxicities. Front Immunol 2020; 11:2023. [PMID: 33123120 PMCID: PMC7572846 DOI: 10.3389/fimmu.2020.02023] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence and continuous development of immune checkpoint inhibitors (ICIs) therapy brings a revolution in cancer therapy history, but the major hurdle associated with their usage is the concomitant ICIs-related toxicities that present a challenge for oncologists. The toxicities may involve non-specific symptoms of multiple systems as for the unique mechanism of formation, which are not easily distinguishable from traditional toxicities. A few of these adverse events are self-limiting and readily manageable, but others may limit treatment, cause interruption and need to be treated with methylprednisolone or tumor necrosis factor-α (TNF-α) antibody infliximab, and even directly threaten life. Early accurate recognition and adequate management are critical to the patient's prognosis and overall survival (OS). Several biomarkers such as the expression of programmed cell death ligand 1 (PD-L1), tumor mutation burden (TMB), and microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) have been proved to be the predictors for anti-tumor efficacy of ICIs, but there is a gap in clinical needs for effective biomarkers that predict toxicities and help filter out the patients who may benefit most from these costly therapies while avoiding major risks of toxicities. Here, we summarize several types of risk factors correlated with ICIs-related toxicities to provide a reference for oncologists to predict the occurrence of ICIs-related toxicities resulting in a timely process in clinical practice.
Collapse
Affiliation(s)
- Ya Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Fu
- Department of Oncology, Xiangyang Hospital, Hubei University of Chinese Medicine, Xiangyang, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Bicheng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
107
|
Bomze D, Asher N, Hasan Ali O, Flatz L, Azoulay D, Markel G, Meirson T. Survival-Inferred Fragility Index of Phase 3 Clinical Trials Evaluating Immune Checkpoint Inhibitors. JAMA Netw Open 2020; 3:e2017675. [PMID: 33095247 PMCID: PMC7584930 DOI: 10.1001/jamanetworkopen.2020.17675] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE In science and medical research, extreme and dichotomous conclusions may be drawn based on whether the P value falls above or below the threshold. The fragility index (ie, the minimum number of changes from nonevents to events resulting in loss of statistical significance) captures the vulnerability of statistics in trials with binary outcomes. There are a growing number of clinical trials of immune checkpoint inhibitors (ICIs), as well as expanding eligibility for patients to receive them. The robustness of survival outcomes in randomized clinical trials (RCTs) should be evaluated using the fragility index extended to time-to-event data. OBJECTIVE To calculate the fragility of survival data in RCTs evaluating ICIs. DESIGN, SETTING, AND PARTICIPANTS In this cross-sectional study, data on phase 3 prospective RCTs investigating ICIs included in PubMed from inception until January 1, 2020, were extracted. Two- or three-group studies reporting results for overall survival were eligible for the survival-inferred fragility index (SIFI) calculation, which is the minimum number of reassignments of the best survivors from the interventional group to the control group resulting in loss of significance (defined as P < .05 by log-rank test). For nonsignificant results, a negative SIFI was calculated by reversing the direction of reassignment (from the control group to the interventional group). MAIN OUTCOMES AND MEASURES Survival-inferred fragility index. RESULTS A total of 45 phase 3 prospective RCTs (4 of which had 3 groups, for a total of 49 groups) were identified, of which 6 (13%) investigated anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) agents, 25 (56%) investigated anti-programmed cell death 1 (PD-1) agents, 12 (27%) investigated anti-programmed cell death 1 ligand 1 agents, and 3 (7%) investigated the combination of anti-CTLA-4 and anti-PD-1 agents. The median SIFI was 5 (interquartile range, -4 to 12) for the intention-to-treat analysis; for these trials, the SIFI was 1% or less of the total sample size in 17 of 49 populations (35%). In 25 of the 49 intention-to-treat populations (51%), the SIFI was less than the number of censored patients in the intervention group shortly after randomization (defined as <5% of the follow-up time). CONCLUSIONS AND RELEVANCE This study suggests that many phase 3 RCTs evaluating ICI therapies have a low SIFI for overall survival, resulting in uncertainty regarding their potential clinical benefit. Although not a definitive solution for the problems arising from dichotomization, SIFI provides an additional means of assessing and communicating the strength of statistical conclusions.
Collapse
Affiliation(s)
- David Bomze
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Nethanel Asher
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | - Omar Hasan Ali
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Lukas Flatz
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Department of Oncology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Daniel Azoulay
- Center for Liver Diseases, Sheba Medical Center, Ramat-Gan, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tomer Meirson
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
108
|
Gogebakan KC, Berry EG, Geller AC, Sonmez K, Leachman SA, Etzioni R. Strategizing Screening for Melanoma in an Era of Novel Treatments: A Model-Based Approach. Cancer Epidemiol Biomarkers Prev 2020; 29:2599-2607. [PMID: 32958498 DOI: 10.1158/1055-9965.epi-20-0881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Benefit-harm tradeoffs of melanoma screening depend on disease risk and treatment efficacy. We developed a model to project outcomes of screening for melanoma in populations with different risks under historic and novel systemic treatments. METHODS Computer simulation model of a screening program with specified impact on overall and advanced-stage incidence. Inputs included meta-analyses of treatment trials, cancer registry data, and a melanoma risk prediction study RESULTS: Assuming 50% reduction in advanced stage under screening, the model projected 59 and 38 lives saved per 100,000 men under historic and novel treatments, respectively. With 10% increase in stage I, the model projects 2.9 and 4.7 overdiagnosed cases per life saved and number needed to be screened (NNS) equal to 1695 and 2632 under historical and novel treatments. When screening was performed only for the 20% of individuals with highest predicted risk, 34 and 22 lives per 100,000 were saved under historic and novel treatments. Similar results were obtained for women, but lives saved were lower. CONCLUSIONS Melanoma early detection programs must shift a substantial fraction of cases from advanced to localized stage to be sustainable. Advances in systemic therapies for melanoma might noticeably reduce benefits of screening, but restricting screening to individuals at highest risk will likely reduce intervention efforts and harms while preserving >50% of the benefit of nontargeted screening. IMPACT Our accessible modeling framework will help to guide population melanoma screening programs in an era of novel treatments for advanced disease.
Collapse
Affiliation(s)
- Kemal Caglar Gogebakan
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Elizabeth G Berry
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Alan C Geller
- Division of Public Health Practice, Harvard School of Public Health, Boston, Massachusetts
| | - Kemal Sonmez
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Statistics, University of Washington, Seattle, Washington
| |
Collapse
|
109
|
Treatment of Advanced Melanoma: Past, Present and Future. Life (Basel) 2020; 10:life10090208. [PMID: 32948031 PMCID: PMC7556013 DOI: 10.3390/life10090208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/17/2022] Open
Abstract
Therapeutic options for treating advanced melanoma are progressing rapidly. Until six years ago, the regimen for treating advanced melanoma mainly comprised cytotoxic agents such as dacarbazine, and type I interferons. Since 2014, anti-programmed cell death 1 (PD1) antibodies have become recognized as anchor drugs for treating advanced melanoma with or without additional combination drugs such as ipilimumab. In addition, v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) kinase inhibitors in combination with mitogen-activated protein kinase kinase (MEK) inhibitors are among the most promising chemotherapeutic regimens for treating advanced BRAF-mutant melanoma, especially in patients with low tumor burden. Since anti-PD1 antibodies are widely applicable for the treatment of both BRAF wild-type and mutated advanced melanomas, several clinical trials for drugs in combination with anti-PD1 antibodies are ongoing. This review focuses on the development of the anti-melanoma therapies available today, and discusses the clinical trials of novel regimens for the treatment of advanced melanoma.
Collapse
|
110
|
Dyer BA, Feng CH, Eskander R, Sharabi AB, Mell LK, McHale M, Mayadev JS. Current Status of Clinical Trials for Cervical and Uterine Cancer Using Immunotherapy Combined With Radiation. Int J Radiat Oncol Biol Phys 2020; 109:396-412. [PMID: 32942005 DOI: 10.1016/j.ijrobp.2020.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 01/05/2023]
Abstract
Novel therapies combined with radiation continue to be of significant interest in the developmental treatment paradigm of gynecologic cancers. Clinical implementation of immunotherapy in oncology has rapidly changed the treatment landscape, options, paradigm, and outcomes through clinical trials. Immunotherapy has emerged as a therapeutic pillar in the treatment of solid tumors with demonstrable synergistic activity when combined with radiation therapy and chemoradiotherapy by an alteration or enhancement of the immune system. In solid tumors, radiation therapy induces migration of dendritic cells, T cell activation, and proliferation, and increases in tumor-infiltrating lymphocytes. These immunomodulatory effects in conjunction with immune checkpoint blockade are currently under active investigation in the adjuvant, definitive, and metastatic settings. Results from early phase trials demonstrate promising efficacy and overall tolerable toxicity profiles of combined modality treatment. There is significant interest in optimizing the treatment for patients with locally advanced cervical cancer beyond the standard of care-chemoradiation-which has been in place for the last 30 years. The majority of cervical cancer emerges after persistent infection with a high-risk subtype of the human papillomavirus, where viral oncoproteins lead to cellular changes and immortalization. As a result, immune tolerance can develop, resulting in cancer. Knowledge of the mechanism of human papillomavirus-related oncogenesis suggests that immune therapy or checkpoint blockade can reinvigorate an antitumor immune response. Current clinical trials are exploring the therapeutic potential of these approaches. Uterine cancers have been grouped into 4 molecular subclasses by their driver mutations, mutational burden, and copy-number alterations. Of these subgroups, the polymerase epsilon-mutated and microsatellite-unstable may represent up to 40% of endometrial cancers, and they have been shown to be immunogenic. Because of the inherent immunogenicity of these MSI-high tumors, combined immune modulation strategies, including chemotherapy, radiation, and immunotherapy and immune checkpoint inhibitor therapy, are being explored to improve treatment outcomes. In this review, we explore current immunomodulatory and multimodality therapeutic approaches in the treatment of cervical and uterine cancer through ongoing clinical trials investigating the combination of immunotherapy and radiation therapy.
Collapse
Affiliation(s)
- Brandon A Dyer
- Department of Radiation Oncology, University of Washington Medical Center, Seattle, Washington
| | - Christine H Feng
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Ramez Eskander
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Services, University of California San Diego, La Jolla, California
| | - Andrew B Sharabi
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Loren K Mell
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Michael McHale
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Services, University of California San Diego, La Jolla, California
| | - Jyoti S Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California.
| |
Collapse
|
111
|
Tong M, Fang X, Yang J, Wu P, Guo Y, Sun J. Abnormal membrane-bound and soluble programmed death ligand 2 (PD-L2) expression in systemic lupus erythematosus is associated with disease activity. Immunol Lett 2020; 227:96-101. [PMID: 32891685 DOI: 10.1016/j.imlet.2020.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Programmed death ligand (PD-L) 2 and PD-L1 are the second and first ligands, respectively, for programmed cell death-1 protein (PD-1), which is one of the key factors responsible for inhibitory T cell signaling, mediating mechanisms of tolerance and providing immune homeostasis. Studies have shown that PD-1 and its ligand PD-L1 are abnormally expressed in autoimmune diseases, such as rheumatoid arthritis and autoimmune hepatitis, but its other ligand, PD-L2, has rarely been studied. This study analyzed the changes in membrane-bound PD-L2 expression in peripheral blood mononuclear cells and soluble PD-L2 (sPD-L2) levels in the serum of patients with systemic lupus erythematosus (SLE) to explore the relationship between PD-L2 expression with disease activity and related test parameters. Our results showed that membrane-bound PD-L2 expression on monocytes was significantly higher and the sPD-L2 levels were significantly lower in SLE patients than in healthy subjects. Patients with active SLE accompanied by lupus nephritis, joint pain, and clinical manifestations of oral ulcers had relatively low secretion of sPD-L2. In addition, this secretion level was significantly and positively correlated with complement components 3 and 4 (C3/C4). These results suggest that PD-L2 may be a promising biomarker associated with the pathogenesis of SLE.
Collapse
Affiliation(s)
- Min Tong
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Xiaohui Fang
- Laboratory, Suzhou Hospital of Traditional Chinese Medicine, Jiangsu, Suzhou, 215009, PR China
| | - Jie Yang
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Pingping Wu
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Yundi Guo
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China
| | - Jing Sun
- Institute of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, Jiangsu, 215009, PR China.
| |
Collapse
|
112
|
Middleton MR, Hoeller C, Michielin O, Robert C, Caramella C, Öhrling K, Hauschild A. Intratumoural immunotherapies for unresectable and metastatic melanoma: current status and future perspectives. Br J Cancer 2020; 123:885-897. [PMID: 32713938 PMCID: PMC7492252 DOI: 10.1038/s41416-020-0994-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/11/2020] [Accepted: 06/29/2020] [Indexed: 01/04/2023] Open
Abstract
The emergence of human intratumoural immunotherapy (HIT-IT) is a major step forward in the management of unresectable melanoma. The direct injection of treatments into melanoma lesions can cause cell lysis and induce a local immune response, and might be associated with a systemic immune response. Directly injecting immunotherapies into tumours achieves a high local concentration of immunostimulatory agent while minimising systemic exposure and, as such, HIT-IT agents are associated with lower toxicity than systemic immune checkpoint inhibitors (CPIs), enabling their potential use in combination with other therapies. Consequently, multiple HIT-IT agents, including oncolytic viruses, pattern-recognition receptor agonists, injected CPIs, cytokines and immune glycolipids, are under investigation. This review considers the current clinical development status of HIT-IT agents as monotherapy and in combination with systemic CPIs, and the practical aspects of administering and assessing the response to these agents. The future of HIT-IT probably lies in its use in combination with systemic CPIs; data from Phase 2 trials indicate a synergy between HIT-IT and CPIs. Data also suggest that the addition of HIT-IT to a CPI might generate responses in CPI-refractory tumours, thereby overcoming resistance and addressing a current unmet need in unresectable and metastatic melanoma for treatment options following progression after CPI treatment.
Collapse
Affiliation(s)
- Mark R Middleton
- University of Oxford Department of Oncology, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK.
| | - Christoph Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Caroline Robert
- Department of Oncology, Gustave Roussy Cancer Campus, Villejuif, and Paris-Saclay University, Orsay, France
| | - Caroline Caramella
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Axel Hauschild
- Department of Dermatology, Venereology and Allergology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
113
|
Abstract
INTRODUCTION Patients with resected stage III melanoma have a heterogeneous prognosis with an especially high risk of relapse for patients with stage IIIB, IIIC and IIID according to the 2018 classification in AJCC Cancer Staging Manual, 8th edition (AJCC-8). Ipilimumab was the first immune checkpoint inhibitor (ICI) to show prolonged overall survival (OS) but at the cost of high toxicity. Pembrolizumab and nivolumab are inhibitors of programmed cell death 1 (PD-1) and showed prolonged relapse-free survival (RFS) in patients with resected stage III melanoma at high risk of relapse compared to placebo and ipilimumab, respectively. AREAS COVERED The aim of this article is to review the mechanisms of action, pharmacokinetics and safety data of pembrolizumab in resected stage III melanoma and to compare its safety profile to other immune checkpoint inhibitors for the same indication. EXPERT OPINION Pembrolizumab as adjuvant therapy of resected stage III melanoma showed an acceptable safety profile, which is comparable to that in advanced melanoma. However, it caused one death. There is uncertainty about its benefits in AJCC-8 stage IIIA melanoma patients. Additionally, caution is required since OS and long-term safety data are not available yet.
Collapse
Affiliation(s)
- F Pham
- Service de Dermatologie, ImmuCare, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon , Pierre-Bénite, France
| | - S Dalle
- Service de Dermatologie, ImmuCare, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon , Pierre-Bénite, France.,UFR de Médecine Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1 , Lyon, France
| |
Collapse
|
114
|
Arensman MD, Yang XS, Zhong W, Bisulco S, Upeslacis E, Rosfjord EC, Deng S, Abraham RT, Eng CH. Anti-tumor immunity influences cancer cell reliance upon ATG7. Oncoimmunology 2020; 9:1800162. [PMID: 32923161 PMCID: PMC7458662 DOI: 10.1080/2162402x.2020.1800162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/02/2020] [Accepted: 07/14/2020] [Indexed: 01/11/2023] Open
Abstract
Macroautophagy (autophagy) is an essential cellular catabolic process required for survival under conditions of starvation. The role of autophagy in cancer is complex, context-dependent and at times contradictory, as it has been shown to inhibit, promote or be dispensable for tumor progression. In this study, we evaluated the contribution of the immune system to the reliance of tumors on autophagy by depleting autophagy-related 7 (ATG7) in murine tumor cells and grafting into immunocompetent versus immunodeficient hosts. Although loss of ATG7 did not affect tumor growth in vitro or in immunodeficient mice, our studies revealed that cancer cell reliance on autophagy was influenced by anti-tumor immune responses, including those mediated by CD8+ T cells. Furthermore, we provide insights into possible mechanisms by which autophagy disruption can enhance anti-tumor immune responses and suggest that autophagy disruption may further benefit patients with immunoreactive tumors.
Collapse
Affiliation(s)
| | - Xiaoran S. Yang
- Pfizer, Oncology Research & Development, Pearl River, NY, USA
| | - Wenyan Zhong
- Pfizer, Oncology Research & Development, Pearl River, NY, USA
| | | | - Erik Upeslacis
- Pfizer, Oncology Research & Development, Pearl River, NY, USA
| | | | - Shibing Deng
- Pfizer, Oncology Research & Development, San Diego, CA, USA
| | | | | |
Collapse
|
115
|
Neurocognitive Function, Psychosocial Outcome, and Health-Related Quality of Life of the First-Generation Metastatic Melanoma Survivors Treated with Ipilimumab. J Immunol Res 2020; 2020:2192480. [PMID: 32775464 PMCID: PMC7391091 DOI: 10.1155/2020/2192480] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/25/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose To assess neurocognitive function (NCF), psychosocial outcome, health-related quality of life (HRQoL), and long-term effects of immune-related adverse events (irAE) on metastatic melanoma survivors treated with ipilimumab (IPI). Methods Melanoma survivors were identified within two study populations (N = 104), at a single-center university hospital, and defined as patients who were disease-free for at least 2 years after initiating IPI. Data were collected using 4 patient-reported outcome measures, computerized NCF testing, and a semistructured interview at the start and 1-year follow-up. Results Out of 18 eligible survivors, 17 were recruited (5F/12M); median age is 57 years (range 33-86); and median time since initiating IPI was 5.6 years (range 2.1-9.3). The clinical interview revealed that survivors suffered from cancer-related emotional distress such as fear of recurrence (N = 8), existential problems (N = 2), survivor guilt (N = 2), and posttraumatic stress disorder (N = 6). The mean EORTC QLQ-C30 Global Score was not significantly different from the European mean of the healthy population. Nine survivors reported anxiety and/or depression (Hospitalization Depression Scale) during the survey. Seven survivors (41%) reported fatigue (Fatigue Severity Scale). Seven patients (41%) had impairment in NCF; only three out of seven survivors had impairment in subjective cognition (Cognitive Failure Questionnaire). Anxiety, depression, fatigue, and neurocognitive symptoms remained stable at the 1-year follow-up. All cases of skin toxicity (N = 8), hepatitis (N = 1), colitis (N = 3), and sarcoidosis (N = 1) resolved without impact on HRQoL. Three survivors experienced hypophysitis; all suffered from persistent fatigue and cognitive complaints 5 years after onset. One survivor who experienced a Guillain-Barré-like syndrome suffered from persisting depression, fatigue, and impairment in NCF. Conclusion A majority of melanoma survivors treated with IPI continue to suffer from emotional distress and impairment in NCF. Timely detection in order to offer tailored care is imperative, with special attention for survivors with a history of neuroendocrine or neurological irAE. The trial is registered with B.U.N. 143201421920.
Collapse
|
116
|
Iacono D, Vitale MG, Basile D, Pelizzari G, Cinausero M, Poletto E, Pascoletti G, Minisini AM. Immunotherapy for older patients with melanoma: From darkness to light? Pigment Cell Melanoma Res 2020; 34:550-563. [PMID: 32745351 DOI: 10.1111/pcmr.12917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/10/2020] [Accepted: 07/28/2020] [Indexed: 01/12/2023]
Abstract
Approximately 40% of malignant melanomas are diagnosed in patients older than 65 years. Elderly patients with melanoma present clinicopathological features related to a more aggressive biology, and they are often diagnosed with advanced stage of disease. Interestingly, in older patients the immune system can be altered with changes both in the innate system and in the adaptive immune system with the acquisition of a pro-inflammatory and immune suppressive phenotype. Immunotherapy with immune checkpoint inhibitors has reshaped the treatment strategies and prognosis of patients with melanoma, and particularly, older age should not be considered a contraindication for immunotherapy. However, data regarding efficacy and safety of immunotherapy in elderly population are still limited because frail older patients are generally excluded from clinical trials. Recently, real-world data have shed light on similar efficacy and safety of immunotherapy in older population compared with younger counterpart. The aim of the present review was to summarize the available knowledge on the underlying immune system in older patients with a diagnosis of melanoma and the immunotherapeutic approaches in this population.
Collapse
Affiliation(s)
- Donatella Iacono
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Maria Grazia Vitale
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy.,Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Debora Basile
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Giacomo Pelizzari
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Marika Cinausero
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Elena Poletto
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Gaetano Pascoletti
- Department of Oncology, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | | |
Collapse
|
117
|
Ben-Aharon O, Magnezi R, Leshno M, Goldstein DA. Mature Versus Registration Studies of Immuno‐Oncology Agents: Does Value Improve With Time? JCO Oncol Pract 2020; 16:e779-e790. [DOI: 10.1200/jop.19.00725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE: A unique feature of immuno-oncology agents is the potential for durable survival for a subset of patients; however, this benefit usually cannot not be seen in the early published data used for regulatory approval. Value frameworks developed by ASCO and the European Society for Medical Oncology (ESMO) assess the clinical benefit demonstrated in clinical trials. Proven benefit may change with time as more mature data are available. Our objective was to evaluate the impact of mature data for immuno-oncology agents on ASCO and ESMO scores and to examine the concordance of these frameworks using more mature data. METHODS: We reviewed Food and Drug Administration (FDA) approvals for immuno-oncology agents between 2011 and 2017, calculated the ASCO-Net Health Benefit (NHB) score and ESMO-Magnitude of Clinical Benefit Score (MCBS), checked which agents fulfilled the criteria of being rewarded for durable survival, assessed the concordance between models using the Spearman correlation test, and compared the initial results of registration studies with mature follow-up data from the same studies. RESULTS: The FDA approved 27 solid tumor indications for immuno-oncology agents between 2011 and 2017. The correlation between ASCO-NHB score and ESMO-MCBS was high (0.88). Mature follow-up data were available for 13 of these indications, in which 6 studies were found to have improved in the grade of ASCO and/or ESMO value frameworks, whereas 2 cases were downgraded in the scale. CONCLUSION: Despite different approaches, the high concordance between ASCO and ESMO value frameworks indicates that both models reward treatments as beneficial for the same immuno-oncology agents. Mature data with longer follow-up reaffirmed most of the findings found in the evaluation in the initially published registration studies.
Collapse
Affiliation(s)
- Omer Ben-Aharon
- Department of Management, Health System Management Program, Bar-Ilan University, Ramat Gan, Israel
| | - Racheli Magnezi
- Department of Management, Health System Management Program, Bar-Ilan University, Ramat Gan, Israel
| | - Moshe Leshno
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | - Daniel A. Goldstein
- Davidoff Cancer Center, Rabin Medical Center, Petah Tiqva, Israel
- Department of Health Policy and Management, University of North Carolina, Chapel Hill, NC
- Department of Oncology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
118
|
Barbari C, Fontaine T, Parajuli P, Lamichhane N, Jakubski S, Lamichhane P, Deshmukh RR. Immunotherapies and Combination Strategies for Immuno-Oncology. Int J Mol Sci 2020; 21:E5009. [PMID: 32679922 PMCID: PMC7404041 DOI: 10.3390/ijms21145009] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
The advent of novel immunotherapies in the treatment of cancers has dramatically changed the landscape of the oncology field. Recent developments in checkpoint inhibition therapies, tumor-infiltrating lymphocyte therapies, chimeric antigen receptor T cell therapies, and cancer vaccines have shown immense promise for significant advancements in cancer treatments. Immunotherapies act on distinct steps of immune response to augment the body's natural ability to recognize, target, and destroy cancerous cells. Combination treatments with immunotherapies and other modalities intend to activate immune response, decrease immunosuppression, and target signaling and resistance pathways to offer a more durable, long-lasting treatment compared to traditional therapies and immunotherapies as monotherapies for cancers. This review aims to briefly describe the rationale, mechanisms of action, and clinical efficacy of common immunotherapies and highlight promising combination strategies currently approved or under clinical development. Additionally, we will discuss the benefits and limitations of these immunotherapy approaches as monotherapies as well as in combination with other treatments.
Collapse
Affiliation(s)
- Cody Barbari
- OMS Students, School of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine (LECOM), 5000 Lakewood Ranch Blvd, Bradenton, FL 34211, USA; (C.B.); (T.F.)
| | - Tyler Fontaine
- OMS Students, School of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine (LECOM), 5000 Lakewood Ranch Blvd, Bradenton, FL 34211, USA; (C.B.); (T.F.)
| | - Priyanka Parajuli
- Department of Internal Medicine, Southern Illinois University, Springfield, IL 62702, USA;
| | - Narottam Lamichhane
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA;
| | - Silvia Jakubski
- Department of Biostatistics, University of Florida, Gainesville, FL 32611, USA;
| | - Purushottam Lamichhane
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine (LECOM), 4800 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| | - Rahul R. Deshmukh
- School of Pharmacy, Lake Erie College of Osteopathic Medicine (LECOM), 5000 Lakewood Ranch Blvd, Bradenton, FL 34211, USA
| |
Collapse
|
119
|
Liu T, Jin B, Chen J, Wang H, Lin S, Dang J, Li G. Comparative risk of serious and fatal treatment-related adverse events caused by 19 immune checkpoint inhibitors used in cancer treatment: a network meta-analysis. Ther Adv Med Oncol 2020; 12:1758835920940927. [PMID: 32774474 PMCID: PMC7394035 DOI: 10.1177/1758835920940927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This network meta-analysis assessed the comparative risk of grade 3-5 and grade 5 treatment-related adverse events (TRAEs) for immune checkpoint inhibitors (ICIs), either alone or in combination with other modalities, for cancer treatment. METHODS PubMed, Embase, Cochrane Library, Web of Science, and recent predominant oncology congresses were searched for relevant phase II and phase III randomized controlled trials (RCTs). As outcomes, grade 3-5, and grade 5 TRAE outcomes were reported as odds ratios and 95% confidence intervals. RESULTS In 67 RCTs involving 36,422 patients and 19 ICIs, the incidence of grade 3-5 and grade 5 TRAEs was 17.9% and 0.8% with ICI monotherapy and 46.3% and 1.4%, respectively, with combinatorial therapy. Pneumonitis was the most common cause of grade 5 TRAEs following either monotherapy (16.3%) or combinatorial therapy (11.4%). Regarding grade 3-5 TRAEs, atezolizumab + chemotherapy (CT) and antiangiogenic therapy (AT) (atezolizumab + CAT), pembrolizumab + CT, ipilimumab + CT, and atezolizumab + CT were more toxic than any ICI monotherapy, pembrolizumab or nivolumab + radiotherapy (RT), and ICIs dual therapy (durvalumab + tremelimumab and nivolumab + ipilimumab). Tremelimumab, ipilimumab, durvalumab, and pembrolizumab were, however, associated with higher grade 5 TRAEs than combinatorial treatments. Atezolizumab + CAT was the most toxic and nivolumab + RT was the least toxic of combinatorial treatments; among monotherapies, tremelimumab and avelumab were the most and least toxic, respectively. The toxicity ranking changed with type of grade 3-5 TRAEs. CONCLUSIONS Compared with combinatorial therapy, ICI monotherapy caused lower grade 3-5 TRAEs, but some monotherapies resulted in a higher incidence of fatal TRAEs. Atezolizumab + CAT and nivolumab + RT were the most and least toxic of combinatorial treatments, respectively, and tremelimumab and avelumab were the most and least toxic of the monotherapies, respectively.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China Department of Radiation Oncology, Anshan Cancer Hospital, Anshan, China
| | - Bo Jin
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jun Chen
- Department of Radiation Oncology, Shenyang Chest Hospital, Shenyang, China
| | - Hui Wang
- Department of Radiation Oncology, General Hospital of Benxi Iron & Steel Industry Group of Liaoning Health Industry Group, Shenyang, China
| | - Shuiyu Lin
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jun Dang
- Department of Radiation Oncology, The First Hospital of China Medical University, 155 Nanjing Road, Heping District, Shenyang, 110001, China
| | - Guang Li
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
120
|
Rossi E, Schinzari G, Maiorano BA, Indellicati G, Di Stefani A, Pagliara MM, Fragomeni SM, De Luca EV, Sammarco MG, Garganese G, Galli J, Blasi MA, Paludetti G, Scambia G, Peris K, Tortora G. Efficacy of immune checkpoint inhibitors in different types of melanoma. Hum Vaccin Immunother 2020; 17:4-13. [PMID: 32663057 PMCID: PMC7872095 DOI: 10.1080/21645515.2020.1771986] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy can be used for cutaneous, mucosal, uveal and conjunctival melanoma. Nevertheless, we cannot expect the same benefit from checkpoint inhibitors for all the types of melanoma. The different biological features can explain the variable efficacy. The main results obtained with immune checkpoint inhibitors in the various types of melanoma were reviewed.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore , Rome, Italy
| | | | - Giulia Indellicati
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy
| | - Alessandro Di Stefani
- Dermatology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome - Italy
| | - Monica Maria Pagliara
- Ophtalmology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy
| | - Simona Maria Fragomeni
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy
| | | | - Maria Grazia Sammarco
- Ophtalmology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy
| | - Giorgia Garganese
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy.,Ginecology and Breast Care Center, Mater Olbia Hospital , Olbia, Italy
| | - Jacopo Galli
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome, Italy
| | - Maria Antonietta Blasi
- Ophtalmology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy.,Ophtalmology, Università Cattolica del Sacro Cuore , Rome, Italy
| | - Gaetano Paludetti
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore , Rome, Italy
| | - Giovanni Scambia
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy.,Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore , Rome, Italy
| | - Ketty Peris
- Dermatology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome - Italy.,Institute of Dermatology, Università Cattolica del Sacro Cuore , Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome, Italy.,Medical Oncology, Università Cattolica del Sacro Cuore , Rome, Italy
| |
Collapse
|
121
|
Immunotargeted therapy in melanoma: patient, provider preferences, and willingness to pay at an academic cancer center. Melanoma Res 2020; 29:626-634. [PMID: 30688762 PMCID: PMC6887632 DOI: 10.1097/cmr.0000000000000572] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Supplemental Digital Content is available in the text. New melanoma therapies have shifted the expectations of patients and providers. Evaluating the impact of treatment characteristics may enhance shared decision making. A survey, including a discrete choice experiment, was utilized to evaluate perceived trade-offs of different melanoma treatments and to estimate out-of-pocket (OOP) willingness-to-pay (WTP) thresholds (January 2016 to March 2016). Participants included patients with melanoma at Huntsman Cancer Institute and their cancer care providers. Stakeholder focus groups were conducted to identify treatment attributes. Descriptive and comparative statistics and multinomial logit model were used to evaluate responses. Response rates were 41.9% (N = 220) for patients and 37.7% (N = 20) for providers. Immunotherapy and targeted therapy attributes considered important by participants were overall survival, immunotherapy-related side effects, and skin toxicities. Patients and providers had significantly different views of quality-of-life expectations, anxiety toward melanoma, trust to make treatment decisions, sharing concerns about treatment, time to discuss treatment, understanding OOP costs, and willingness to undergo/recommend treatment (half of the patients would undergo treatment if it was effective for > 24 months). Among patients, the average monthly OOP WTP for combination immunotherapy with nivolumab + ipilimumab was $ 2357 and for BRAF/MEK inhibitor was $1648. Among providers, these estimates were $ 2484 and $1350, respectively. Discordance existed between patients’ and providers’ perceptions about quality of life expectations, degree of anxiety, sharing of opinions, and progression-free survival. Our study suggests that patients and providers exhibit a higher OOP WTP for combination immunotherapy treatment compared with BRAF/MEK inhibitors, influenced predominately by overall survival expectations.
Collapse
|
122
|
Bhat TS, Rosman IS, Cornelius LA, Musiek AC. Mycosis fungoides associated with recurrence of malignant melanoma. JAAD Case Rep 2020; 6:793-795. [PMID: 32775589 PMCID: PMC7394829 DOI: 10.1016/j.jdcr.2020.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Trisha S. Bhat
- Division of Dermatology, Washington University School of Medicine, St Louis, Missouri
- Correspondence to: Trisha S. Bhat, BA, Division of Dermatology, Washington University School of Medicine, 660 S. Euclid Ave, Campus Box 8123, St Louis, MO 63110.
| | - Ilana S. Rosman
- Division of Dermatology, Washington University School of Medicine, St Louis, Missouri
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Lynn A. Cornelius
- Division of Dermatology, Washington University School of Medicine, St Louis, Missouri
| | - Amy C. Musiek
- Division of Dermatology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
123
|
Almeida AA, Lima GDA, Simão MVRC, Moreira GA, Siqueira RP, Zanatta AC, Vilegas W, Machado‐Neves M, Bressan GC, Leite JPV. Screening of plants from the Brazilian Atlantic Forest led to the identification of Athenaea velutina (Solanaceae) as a novel source of antimetastatic agents. Int J Exp Pathol 2020; 101:106-121. [PMID: 32452573 PMCID: PMC7370850 DOI: 10.1111/iep.12351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/22/2020] [Accepted: 03/23/2020] [Indexed: 12/26/2022] Open
Abstract
Plant biodiversity is a source of potential natural products for the treatment of many diseases. One of the ways of discovering new drugs is through the cytotoxic screening of extract libraries. The present study evaluated 196 extracts prepared by maceration of Brazilian Atlantic Forest trees with organic solvents and distilled water for cytotoxic and antimetastatic activity. The MTT assay was used to screen the extract activity in MCF-7, HepG2 and B16F10 cancer cells. The highest cytotoxic extract had antimetastatic activity, as determined in in vitro assays and melanoma murine model. The organic extract of the leaves of Athenaea velutina (EAv) significantly inhibited migration, adhesion, invasion and cell colony formation in B16F10 cells. The phenolic compounds and flavonoids in EAv were identified for the first time, using flow injection with electrospray negative ionization-ion trap tandem mass spectrometry analysis (FIA-ESI-IT-MSn ). EAv markedly suppressed the development of pulmonary melanomas following the intravenous injection of melanoma cells to C57BL/6 mice. Stereological analysis of the spleen cross-sections showed enlargement of the red pulp area after EAv treatment, which indicated the activation of the haematopoietic system. The treatment of melanoma-bearing mice with EAv did not result in liver damage. In conclusion, these findings suggest that A velutina is a source of natural products with potent antimetastatic activity.
Collapse
Affiliation(s)
- Alisson A. Almeida
- Departamento de Bioquímica e Biologia MolecularUniversidade Federal de ViçosaMinas GeraisBrazil
| | - Graziela D. A. Lima
- Departamento de Biologia GeralUniversidade Federal de ViçosaMinas GeraisBrazil
| | | | - Gabriela A. Moreira
- Departamento de Bioquímica e Biologia MolecularUniversidade Federal de ViçosaMinas GeraisBrazil
| | - Raoni P. Siqueira
- Departamento de Bioquímica e Biologia MolecularUniversidade Federal de ViçosaMinas GeraisBrazil
| | | | | | | | - Gustavo C. Bressan
- Departamento de Bioquímica e Biologia MolecularUniversidade Federal de ViçosaMinas GeraisBrazil
| | - João P. V. Leite
- Departamento de Bioquímica e Biologia MolecularUniversidade Federal de ViçosaMinas GeraisBrazil
| |
Collapse
|
124
|
Golay J, Andrea AE. Combined Anti-Cancer Strategies Based on Anti-Checkpoint Inhibitor Antibodies. Antibodies (Basel) 2020; 9:E17. [PMID: 32443877 PMCID: PMC7345008 DOI: 10.3390/antib9020017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Therapeutic monoclonal antibodies for the treatment of cancer came of age in 1997, with the approval of anti-CD20 Rituximab. Since then, a wide variety of antibodies have been developed with many different formats and mechanisms of action. Among these, antibodies blocking immune checkpoint inhibitors (ICI) have revolutionized the field, based on the novelty of their concept and their demonstrated efficacy in several types of cancer otherwise lacking effective immunotherapy approaches. ICI are expressed by tumor, stromal or immune cells infiltrating the tumor microenvironment, and negatively regulate anti-tumor immunity. Antibodies against the first discovered ICI, CTLA-4, PD-1 and PD-L1, have shown significant activity in phase III studies against melanoma and other solid cancers, alone or in combination with chemotherapy or radiotherapy. However, not all cancers and not all patients respond to these drugs. Therefore, novel antibodies targeting additional ICI are currently being developed. In addition, CTLA-4, PD-1 and PD-L1 blocking antibodies are being combined with each other or with other antibodies targeting novel ICI, immunostimulatory molecules, tumor antigens, angiogenic factors, complement receptors, or with T cell engaging bispecific antibodies (BsAb), with the aim of obtaining synergistic effects with minimal toxicity. In this review, we summarize the biological aspects behind such combinations and review some of the most important clinical data on ICI-specific antibodies.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy “G. Lanzani”, UOC Ematologia, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy
- Fondazione per la Ricerca Ospedale Maggiore, 24127 Bergamo, Italy
| | - Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut 1100, Lebanon;
| |
Collapse
|
125
|
Cofano F, Monticelli M, Ajello M, Zenga F, Marengo N, Di Perna G, Altieri R, Cassoni P, Bertero L, Melcarne A, Tartara F, Ducati A, Garbossa D. The Targeted Therapies Era Beyond the Surgical Point of View: What Spine Surgeons Should Know Before Approaching Spinal Metastases. Cancer Control 2020; 26:1073274819870549. [PMID: 31865766 PMCID: PMC6728684 DOI: 10.1177/1073274819870549] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last few years, the treatment of spinal metastases has significantly
changed. This is due to the advancements in surgical technique, radiotherapy,
and chemotherapy which have enriched the multidisciplinary management. Above
all, the field of molecular biology of tumors is in continuous and prosperous
evolution. In this review, the molecular markers and new approaches that have
radically modified the chemotherapeutic strategy of the most common metastatic
neoplasms will be examined together with clinical and surgical implications. The
experience and skills of several different medical professionals are mandatory:
an interdisciplinary oncology team represents the winning strategy in the
treatment of patients with spinal metastases
Collapse
Affiliation(s)
- Fabio Cofano
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Matteo Monticelli
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Marco Ajello
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Francesco Zenga
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Nicola Marengo
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Giuseppe Di Perna
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Roberto Altieri
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Paola Cassoni
- Dipartimento di Scienze Mediche, Pathology, Universita degli Studi di Torino, Torino, Italy
| | - Luca Bertero
- Dipartimento di Scienze Mediche, Pathology, Universita degli Studi di Torino, Torino, Italy
| | - Antonio Melcarne
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Fulvio Tartara
- Azienda Ospedaliero-Universitaria di Parma, Special surgery, Neurosurgery, Torino, Italy
| | - Alessandro Ducati
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| | - Diego Garbossa
- Dipartimento di Neuroscienze, Neurosurgery, Universita degli Studi di Torino, Torino, Italy
| |
Collapse
|
126
|
De Luca R, Meraviglia S, Blasi L, Maiorana A, Cicero G. Nivolumab in metastatic melanoma: good efficacy and tolerability in elderly patients. ACTA ACUST UNITED AC 2020; 27:e75-e80. [PMID: 32489255 DOI: 10.3747/co.27.5293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Nivolumab is an anti-PD-1 antibody that restores the antitumour immune function of T cells, blocking the binding of PD-1 with its ligand PD-L1. PD-1 is expressed on T cells and interacts with PD-L1 on tumour cells. The PD-1-PD-L1 link inhibits T cell activation. In metastatic melanoma, PD-1-PD-L1 binding plays a critical role, and the advent of the immune checkpoint inhibitor nivolumab has delivered new and effective treatment options with proven clinical benefit. In the present study, we evaluated the efficacy of nivolumab in elderly patients with metastatic melanoma. Methods The study enrolled 55 elderly patients (75 years of age and older) with a diagnosis of metastatic melanoma. Primary endpoints of the study were progression-free survival (pfs) and the objective response rate; secondary endpoints were overall survival, reduction in serum lactate dehydrogenase (ldh) from before to after treatment, and tolerability. Results Nivolumab was well tolerated and resulted in good disease control, with a manageable toxicity profile and significant clinical benefit. The duration of pfs was 5.1 months (95% confidence interval: 3.5 months to 6.8 months). A significant correlation was observed between reduction in serum ldh and pfs: 0.60 (95% confidence interval: 0.28 to 0.86; p = 0.002). Conclusions Nivolumab is an immunotherapy treatment that has proved to be an effective and well-tolerated therapeutic option in elderly patients with metastatic melanoma.
Collapse
Affiliation(s)
- R De Luca
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - S Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy
| | - L Blasi
- Medical Oncology Unit, arnas Ospedali Civico Di Cristina Benfratelli, Palermo, Italy
| | - A Maiorana
- Department of Surgical, Oncological and Oral Sciences, Section of Surgical Oncology, University of Palermo, Palermo, Italy
| | - G Cicero
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
127
|
Ugurel S, Röhmel J, Ascierto PA, Becker JC, Flaherty KT, Grob JJ, Hauschild A, Larkin J, Livingstone E, Long GV, Lorigan P, McArthur GA, Ribas A, Robert C, Zimmer L, Schadendorf D, Garbe C. Survival of patients with advanced metastatic melanoma: The impact of MAP kinase pathway inhibition and immune checkpoint inhibition - Update 2019. Eur J Cancer 2020; 130:126-138. [DOI: 10.1016/j.ejca.2020.02.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/02/2020] [Indexed: 12/19/2022]
|
128
|
Abstract
OPINION STATEMENT With greater understanding of underlying biology and development of effective BRAF-targeted therapy and immunotherapy, along with remarkable advances in local treatment such as stereotactic radiosurgery, melanoma brain metastasis (MBM) is witnessing continually improving outcome, with 1-year overall survival rate approaching 85%. Given disease complexity and myriad treatment options, all patients with MBM should ideally be evaluated in a multidisciplinary setting to allow an individualized treatment approach based on prognostic groups, molecular classification, number and size of brain metastasis, and performance status. With improving outcome, pendulum has now swayed to focus more on effective treatment modalities with minimal neurological toxicity while maintaining quality of life. Surgery is usually considered in symptomatic and large MBMs, while stereotactic radiosurgery considered in 1-4 lesions, and now also being explored for up to 15 brain metastases for improved local control. The role of whole brain radiotherapy is diminishing given its neurocognitive toxicities and is reserved for patients with diffuse brain involvement. Cytotoxic chemotherapy has largely been ineffective without evidence for survival benefit. Immune checkpoint inhibitors have become the cornerstone of management for melanoma brain metastasis with durable intracranial tumor control and excellent toxicity profile. For patients with asymptomatic MBMs, ipilimumab and nivolumab have shown intracranial response near 60% and provides comparable clinical benefit in MBMs as for extracranial metastases. For patients with driver BRAF mutation, BRAFi-/MEKi-targeted agents are proven to be effective in MBM with high rate intracranial responses (44-59%). However, the durability of intracranial responses induced by BRAFi/MEKi seems to be shorter than that of extracranial disease. Emerging data support novel combination of systemic therapy and stereotactic radiosurgery, which appears to be safe and effective; however, potential benefits and risks should be evaluated prospectively. Promising ongoing trials will further expand therapeutic evidence in MBM, and patients should be encouraged to participate in clinical trials.
Collapse
Affiliation(s)
- Anupam Rishi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, 33612, USA
| | - Hsiang-Hsuan Michael Yu
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, 33612, USA.
| |
Collapse
|
129
|
Amaral T, Seeber O, Mersi E, Sanchez S, Thomas I, Meiwes A, Forschner A, Leiter U, Eigentler T, Keim U, Garbe C. Primary Resistance to PD-1-Based Immunotherapy-A Study in 319 Patients with Stage IV Melanoma. Cancers (Basel) 2020; 12:cancers12041027. [PMID: 32331243 PMCID: PMC7226601 DOI: 10.3390/cancers12041027] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Primary resistance to immunotherapy can be observed in approximately 40–65% of the stage IV melanoma patients treated with immune checkpoint inhibitors. A minority of the patients receive a second-line therapy, and the clinical benefit is small. Patients and methods: Stage IV melanoma patients treated with first-line PD-1-based immunotherapy between January 2015 and December 2018 were investigated. Primary resistance was defined as progressive disease (PD) at the time of the first tumor assessment after starting immunotherapy. Patients with complete response, partial response, and stable disease were classified as having disease control (DC). Overall survival (OS) and progression-free survival (PFS) were evaluated by Kaplan–Meier estimator. Univariate and multivariate logistic regression analyses were performed to determine prognostic factors associated with OS. Results: Three hundred and nineteen patients were included, and 40% had primary resistance to immunotherapy. The median follow-up time was 22 months. Patients with primary resistance had 1-, 2-, and 3-year OS rates of 41%, 15%, and 10%, respectively, compared to 91%, 81%, and 65% for the patients who achieved DC. The following independently significant prognostic factors for OS were identified: protein S100B level and primary tumor localization. There was a statistically significant difference for OS (p < 0.0001) but not for PFS (p = 0.230) when analyzing risk groups formed with a combination of these two variables (low-, intermediate-, and high-risk subgroups). Conclusions: Melanoma patients with primary resistance to immunotherapy have a dismal prognosis. Response at the first tumor assessment after starting immunotherapy is a stronger prognostic factor for the further course of the disease than pretreatment risk factors.
Collapse
Affiliation(s)
- Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
- Portuguese Air Force—Health Care Direction, 1649-020 Lisbon, Portugal
| | - Olivia Seeber
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Edgar Mersi
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Stephanie Sanchez
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ioannis Thomas
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Andreas Meiwes
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Thomas Eigentler
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ulrike Keim
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
- Correspondence: ; Tel.: +49-7071-298-87110; Fax: +49-7071-29-51-87
| |
Collapse
|
130
|
Jacqueline C, Finn OJ. Antibodies specific for disease-associated antigens (DAA) expressed in non-malignant diseases reveal potential new tumor-associated antigens (TAA) for immunotherapy or immunoprevention. Semin Immunol 2020; 47:101394. [PMID: 32273212 DOI: 10.1016/j.smim.2020.101394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune responses to a large number of mutated and non-mutated tumor antigens have been studied in an attempt to unravel the highly complex immune response to cancer. Better understanding of both the effectors and the targets of successful immunosurveillance can inform various immunotherapeutic approaches, which can strengthen or replace natural immunosurveillance that a tumor has managed to escape. In this review we highlight targets of antibodies generated in the context of diseases other than cancer, such as asthma, allergies, autoimmune disorders, inflammation and infections, where the antibody presence correlates either with an increased or a reduced lifetime risk of cancer. We focus on their target antigens, self-molecules abnormally expressed on diseased cells or cross-reactive with exogenous antigens and found on cancer cells as tumor associated antigens (TAA). We refer to them as disease-associated antigens (DAA). We review 4 distinct categories of antibodies according to their target DAA, their origin and their reported impact on cancer risk: natural antibodies, autoantibodies, long-term memory antibodies and allergy-associated antibodies. Increased understanding and focus on their specific targets could enable a more rational choice of antigens for both therapeutic and preventative cancer vaccines and other more effective and less toxic cancer immunotherapies.
Collapse
Affiliation(s)
- Camille Jacqueline
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
131
|
Karyopherin α 2 promotes proliferation, migration and invasion through activating NF-κB/p65 signaling pathways in melanoma cells. Life Sci 2020; 252:117611. [PMID: 32243925 DOI: 10.1016/j.lfs.2020.117611] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/12/2020] [Accepted: 03/28/2020] [Indexed: 12/18/2022]
Abstract
AIMS Melanoma is a fatal malignancy. Karyopherin α 2 (KPNA2) plays an important role in many carcinogenesis. This study was aimed to study the role of KPNA2 in cellular functions and molecular mechanisms of melanoma. MAIN METHODS We investigated the expression and prognosis of KPNA2 in melanoma using the GEPIA database (http://gepia.cancer-pku.cn/). The effect of KPNA2 on melanoma cells was determined using real-time PCR, western blot, immunofluorescence assay, CCK-8, colony formation, wound healing assay, transwell assay, EMSA, and immunohistochemistry. The influence of KPNA2 on the tumorigenicity of melanoma cells was evaluated in a nude mice model in vivo. KEY FINDINGS Our results showed that KPNA2 expression is relatively high in melanoma tissues and cells, and melanoma patients with higher expression of KPNA2 had lower overall survival rate and disease free survival rate. KPNA2 promoted proliferation ability and increased the expression of PCNA, Ki67, and C-MYC in melanoma cells. Further, KPNA2 could promote migration and invasion and increase the expression of MMP2 and MMP9. Mechanism studies showed that KPNA2 activated NF-κB/p65 signaling pathways, as evidenced by the nuclear translocation of p65 and increased the expression of COX-2, ICAM-1, iNOS, and MCP1 in melanoma cells. NF-κB inhibitor JSH-23 could reverse the pro-tumor effects of KPNA2 on melanoma cells. Moreover, upregulation of KPNA2 facilitated the tumorigenicity of melanoma cells. SIGNIFICANCE KPNA2 promotes proliferation, migration and invasion through enhancing NF-κB/p65 signaling pathways in melanoma cells. Our study suggests KPNA2 as a potential therapeutic target for the treatment of melanoma.
Collapse
|
132
|
Dragomir MP, Kopetz S, Ajani JA, Calin GA. Non-coding RNAs in GI cancers: from cancer hallmarks to clinical utility. Gut 2020; 69:748-763. [PMID: 32034004 DOI: 10.1136/gutjnl-2019-318279] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/10/2019] [Accepted: 12/14/2019] [Indexed: 12/11/2022]
Abstract
One of the most unexpected discoveries in molecular oncology, in the last decades, was the identification of a new layer of protein coding gene regulation by transcripts that do not codify for proteins, the non-coding RNAs. These represent a heterogeneous category of transcripts that interact with many types of genetic elements, including regulatory DNAs, coding and other non-coding transcripts and directly to proteins. The final outcome, in the malignant context, is the regulation of any of the cancer hallmarks. Non-coding RNAs represent the most abundant type of hormones that contribute significantly to cell-to cell communication, revealing a complex interplay between tumour cells, tumour microenvironment cells and immune cells. Consequently, profiling their abundance in bodily fluids became a mainstream of biomarker identification. Therapeutic targeting of non-coding RNAs represents a new option for clinicians that is currently under development. This review will present the biology and translational value of three of the most studied categories on non-coding RNAs, the microRNAs, the long non-coding RNAs and the circular RNAs. We will also focus on some aspirational concepts that can help in the development of clinical applications related to non-coding RNAs, including using pyknons to discover new non-coding RNAs, targeting human-specific transcripts which are expressed specifically in the tumour cell and using non-coding RNAs to increase the efficiency of immunotherapy.
Collapse
Affiliation(s)
- Mihnea Paul Dragomir
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - George Adrian Calin
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
133
|
Kaštelan S, Antunica AG, Oresković LB, Pelčić G, Kasun E, Hat K. Immunotherapy for Uveal Melanoma - Current Knowledge and Perspectives. Curr Med Chem 2020; 27:1350-1366. [DOI: 10.2174/0929867326666190704141444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 06/09/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022]
Abstract
Uveal melanoma is the most prevalent primary intraocular tumour in adults with
the incidence between five and six cases per million people in the United States and Europe.
The prognosis of patients with uveal melanoma is unfavourable with a 5-year survival rate of
50-70% despite significant advances in local tumour treatment using radiotherapy or surgical
resection. Approximately 50% of the patients develop metastases within 15 years from initial
diagnosis, mostly in the liver. The median survival rate after the onset of metastases is 6
months. Potential treatment options for metastatic uveal melanoma are chemotherapy, targeted
therapy, and immunotherapy but no method showed satisfactory results. Immunotherapy
with checkpoint inhibition showed promising results in the treatment of cutaneous melanoma;
however, it did not appear to be equally effective with uveal melanoma. This may be
due to differences in mutational burden, expression of neoantigens between these two types of
tumour, immunosuppressive tumour microenvironment, and low immunogenicity and immune
privilege of uveal melanoma. Considering the disappointing results of treatment with
anti-CTLA-4 and PD-1/PD-L1 blockade in patients with advanced uveal melanoma several
new forms of therapies are being developed. This may include immunotherapy with
IMCgp100, glembatumumab vedotin and the infusion of autologous TILs, targeted therapy
with selective MEK inhibitors, epigenetic therapy, and nanotherapy. Better insight into the
molecular and genetic profile of uveal melanoma will facilitate detection of new prognostic
biomarkers and thus enable a better modification of the existing immunotherapy methods and
development of new forms of treatment specifically designed for uveal melanoma patients.
Collapse
Affiliation(s)
- Snježana Kaštelan
- Department of Ophthalmology, University Hospital Dubrava, Zagreb, Croatia
| | | | | | - Goran Pelčić
- Department of Ophthalmology, Faculty of Medicine, University of Rijeka and Clinical Hospital Center Rijeka, Rijeka, Croatia
| | - Ema Kasun
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Koraljka Hat
- Department of Maxillofacial Surgery, University Hospital Dubrava, Zagreb, Croatia
| |
Collapse
|
134
|
Clinical Update on Checkpoint Inhibitor Therapy for Conjunctival and Eyelid Melanoma. Int Ophthalmol Clin 2020; 60:77-89. [PMID: 32205655 DOI: 10.1097/iio.0000000000000308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
135
|
Krattinger R, Ramelyte E, Dornbierer J, Dummer R. Is single versus combination therapy problematic in the treatment of cutaneous melanoma? Expert Rev Clin Pharmacol 2020; 14:9-23. [PMID: 31364890 DOI: 10.1080/17512433.2019.1650641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: The development of immunotherapies and targeted therapies has changed the treatment approach in resectable, nonresectable, and metastatic melanoma. Because of their different pharmacological profiles, immunotherapies and/or targeted therapies have been studied in various combinations. Areas covered: We reviewed PubMed for most important clinical trials investigating efficacy and tolerability of combinatorial and single-agent approaches for the treatment of melanoma that were published up to June 2019. We discuss the most promising therapy approaches and highlight challenges of melanoma treatment. Expert opinion: Combinatorial approaches seem to be very promising in the treatment of resectable and advanced melanoma. Currently, dual immune checkpoint inhibition (ICI) with nivolumab and ipilimumab offers the best first-line treatment option for patients with BRAF-wt and -mutated, advanced melanoma. It is therapy of choice in younger patients with good ECOG performance status and poor prognostic features, whereas ICI monotherapy should be preferred in elderly patients with advanced melanoma. Benefit-risk ratio, patient's QoL and expectations, as well as treatment costs have to be considered in the choice of treatment. However, to elucidate mechanisms of resistance, biomarkers of response and to better define personalized strategies in the treatment of cutaneous melanoma, larger clinical trials comparing combined versus sequential therapies are necessary.
Collapse
Affiliation(s)
- Regina Krattinger
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Joëlle Dornbierer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| |
Collapse
|
136
|
Current Advances in the Treatment of BRAF-Mutant Melanoma. Cancers (Basel) 2020; 12:cancers12020482. [PMID: 32092958 PMCID: PMC7072236 DOI: 10.3390/cancers12020482] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most lethal form of skin cancer. Melanoma is usually curable with surgery if detected early, however, treatment options for patients with metastatic melanoma are limited and the five-year survival rate for metastatic melanoma had been 15-20% before the advent of immunotherapy. Treatment with immune checkpoint inhibitors has increased long-term survival outcomes in patients with advanced melanoma to as high as 50% although individual response can vary greatly. A mutation within the MAPK pathway leads to uncontrollable growth and ultimately develops into cancer. The most common driver mutation that leads to this characteristic overactivation in the MAPK pathway is the B-RAF mutation. Current combinations of BRAF and MEK inhibitors that have demonstrated improved patient outcomes include dabrafenib with trametinib, vemurafenib with cobimetinib or encorafenib with binimetinib. Treatment with BRAF and MEK inhibitors has met challenges as patient responses began to drop due to the development of resistance to these inhibitors which paved the way for development of immunotherapies and other small molecule inhibitor approaches to address this. Resistance to these inhibitors continues to push the need to expand our understanding of novel mechanisms of resistance associated with treatment therapies. This review focuses on the current landscape of how resistance occurs with the chronic use of BRAF and MEK inhibitors in BRAF-mutant melanoma and progress made in the fields of immunotherapies and other small molecules when used alone or in combination with BRAF and MEK inhibitors to delay or circumvent the onset of resistance for patients with stage III/IV BRAF mutant melanoma.
Collapse
|
137
|
Buyse M, Saad ED, Burzykowski T, Péron J. Assessing Treatment Benefit in Immuno-oncology. STATISTICS IN BIOSCIENCES 2020. [DOI: 10.1007/s12561-020-09268-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
138
|
Wang YB, Lv G, Xu FH, Ma LL, Yao YM. Comprehensive Survey of Clinical Trials Registration for Melanoma Immunotherapy in the ClinicalTrials.gov. Front Pharmacol 2020; 10:1539. [PMID: 31998135 PMCID: PMC6966167 DOI: 10.3389/fphar.2019.01539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Comprehensively evaluate the immunotherapeutic clinical trials and provide reference for melanoma treatment and research. Methods: The website of ClinicalTrials.gov was searched to retrieve and download all registered clinical trials for melanoma immunotherapy on August 1 (updated on August 25), 2019. All registration trials met the inclusion criteria were collected regardless of the type of study, the status of recruitment, and the results of the study. The general characteristics, methodological characteristics, and the types of immunotherapeutic drugs included of these trials were analyzed. Results: Finally, 242 eligible trials were included and evaluated. Of them, 30.6% were completed, 16.9% were terminated, and two were withdrawn; 77.7% recruited less than 100 participants; 30.5% were randomized; 45.5% was single group assignment; 88.8% were not masked; the primary purpose was treatment; 44.2% had data on monitoring committees; 27.7% used US FDA-regulated immunization drugs; 78.5% without results posted; 43.0% were sponsored by the industry. Immunological checkpoint inhibitors were most often studied, with 53.6% of the trials involving PD-1, the most commonly studied was Nivolumab. Conclusions: Currently, most of the registered clinical trials for melanoma immunotherapy were interventional open-label trials. Most immunotherapy research hotspots were in the FDA-regulated drug product, and a few trials reported available test results. It is necessary to strengthen the supervision of results and explore and disseminate more effective and safe immunotherapy methods.
Collapse
Affiliation(s)
- Yan-Bo Wang
- Department of Microbiology and Immunology, Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Gang Lv
- Department of General Surgery, The 8th Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Feng-Hua Xu
- Ward I of Internal Medicine, Beijing General Hospital of the Chinese People's Armed Police Force, Beijing, China
| | - Lin-Lu Ma
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yong-Ming Yao
- Department of Microbiology and Immunology, Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
139
|
Flem-Karlsen K, Nyakas M, Farstad IN, McFadden E, Wernhoff P, Jacobsen KD, Flørenes VA, Mælandsmo GM. Soluble AXL as a marker of disease progression and survival in melanoma. PLoS One 2020; 15:e0227187. [PMID: 31917795 PMCID: PMC6952099 DOI: 10.1371/journal.pone.0227187] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022] Open
Abstract
Receptor tyrosine kinase AXL is a one-pass transmembrane protein upregulated in cancers and associated with lower survival and therapy resistance. AXL can be cleaved by the A Disintegrin and Metalloproteinases (ADAM)10 and ADAM17, yielding a soluble version of the protein. Elevated soluble AXL (sAXL) has been reported to be associated with disease progression in hepatocellular carcinoma, renal cancer, neurofibromatosis type 1 and inflammatory diseases. In the present work, we analyzed sAXL levels in blood from melanoma patients and showed that sAXL increases with disease progression. Additionally, increased sAXL levels were found correlated with shorter two-year survival in stage IV patients treated with ipilimumab. Furthermore, we showed that sAXL levels were related to the percentage of cells expressing AXL in resected melanoma lymph node metastases. This finding was verified in vitro, where sAXL levels in the cell media corresponded to AXL expression in the cells. AXL inhibition using the small-molecular inhibitor BGB324 reduced sAXL levels, while the cellular expression was elevated through increased protein stability. Our findings signify that quantification of sAXL blood levels is a simple and easily assessable method to determine cellular AXL levels and should be further evaluated for its use as a biomarker of disease progression and treatment response.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Marta Nyakas
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Inger Nina Farstad
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Erin McFadden
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Patrik Wernhoff
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kari Dolven Jacobsen
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Vivi Ann Flørenes
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute of Medical Biology, Faculty of Health Sciences, UiT–Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
140
|
Dubois M, Ardin C, André F, Scherpereel A, Mortier L. [The revolution of immuno-oncology therapy: review of immune checkpoint inhibitors efficacy]. Med Sci (Paris) 2020; 35:937-945. [PMID: 31903897 DOI: 10.1051/medsci/2019225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Immunotherapy represents a major paradigm shift, as the treatment no longer directly targets tumor cells, but the patient him/herself, in order to restore an effective anti-tumor immunity. This article illustrates the growing place of immune checkpoint inhibitors in the available therapeutic options, by focusing on two cancers with poor outcome: metastatic melanoma and metastatic non-small cell lung cancer (NSCLC), against which Immune checkpoints inhibitors now occupy a central place. Many questions remain unresolved, such as the search for markers predicting a good response to treatment, which would allow the selection of responder patients. Numerous trials are in progress, evaluating the relevance of these new molecules at earlier stages of the disease (adjuvant and neoadjuvant strategies) and their place in combined strategies (associated with chemotherapy, targeted therapies, and other types of immunotherapy).
Collapse
Affiliation(s)
- Manon Dubois
- Service de dermatologie, CHU Lille, Lille, France
| | - Camille Ardin
- Service de pneumologie et oncologie thoracique, CHU Lille, Lille, France
| | - Fanny André
- Service de dermatologie, CHU Lille, Lille, France
| | - Arnaud Scherpereel
- Service de pneumologie et oncologie thoracique, CHU Lille, Lille, France - Université de Lille, Lille, France - Unité Inserm 1189 OncoThAI, Lille, France
| | - Laurent Mortier
- Service de dermatologie, CHU Lille, Lille, France - Université de Lille, Lille, France - Unité Inserm 1189 OncoThAI, Lille, France
| |
Collapse
|
141
|
Paschen A, Schadendorf D. The Era of Checkpoint Inhibition: Lessons Learned from Melanoma. Recent Results Cancer Res 2020; 214:169-187. [PMID: 31473853 DOI: 10.1007/978-3-030-23765-3_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Treatment of patients with advanced metastatic melanoma has for decades been a story of very limited success. This dramatically changed when therapy with anti-PD-1 checkpoint blocking antibodies was approved in the USA and Europe in 2014 and 2015, respectively. The therapy exploits the capacity of CD8+ T cells to specifically kill tumor cells. Within the tumor microenvironment, CD8+ T cell activity is blocked by suppressive signals received via PD-1, an inhibitory co-receptor and so-called checkpoint of T cell activation. PD-1 binds to its ligand PD-L1 on melanoma cells which dampens the T cell's activity. Antibodies blocking inhibitory PD-1/PD-L1 interaction release T cells from suppression. Treatment of late-stage disease melanoma patients with antibodies targeting the PD-1/PD-L1 axis, termed immune checkpoint blocking therapy (ICBT), yields clinical frequently long-lasting responses in 30-40% of cases. Despite this remarkable breakthrough, still the majority of patients resists ICBT or develops resistance after initial therapy response. Administration of anti-PD-1 antibodies in combination with antibodies targeting CTLA-4, another inhibitory immune checkpoint increased clinical responses rate up to 50% but at costs of higher treatment-related toxicities. Thus, strong efforts are now directed toward the understanding of therapy resistance, the identification of biomarkers predicting therapy response, and the development of alternative PD-1-based combination treatment to improve patient outcomes.
Collapse
Affiliation(s)
- Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
142
|
Dan H, Zhang S, Zhou Y, Guan Q. DNA Methyltransferase Inhibitors: Catalysts For Antitumour Immune Responses. Onco Targets Ther 2019; 12:10903-10916. [PMID: 31849494 PMCID: PMC6913319 DOI: 10.2147/ott.s217767] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Epigenetics is a kind of heritable change that involves the unaltered DNA sequence and can have effects on gene expression. The regulatory mechanism mainly includes DNA methylation, histone modification and non-coding RNA regulation. DNA methylation is currently the most studied aspect of epigenetics. It is widely present in eukaryotic cells and is the most important epigenetic mark in the regulation of gene expression in the cell. DNA methyltransferase inhibitors (DNMTi) have been increasingly recognized in the field of cancer immunotherapy, have been approved for the treatment of acute myeloid leukaemia (AML) and are widely being used in clinical trials of cancer immunotherapies. DNMTi promote the reactivation of tumour suppressor genes, enhance tumour immunogenicity, and stimulate a variety of immune cells to secrete cytokines that exert cytotoxic effects, promote tumour cell death, including macrophages, natural killer (NK) cells and CD8+ T cells, and upregulate major histocompatibility complex (MHC) class I expression levels. Here, we mainly summarize the epigenetics related to DNMTi and their regulation of the antitumour immune response and DNMTi combined with immuno-therapeutics or histone deacetylase inhibitors to demonstrate the great development potential and clinical application value of DNMTi.
Collapse
Affiliation(s)
- Huimin Dan
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Shanshan Zhang
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Yongning Zhou
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| | - Quanlin Guan
- Gansu Province Key Laboratory of Gastrointestinal Diseases, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu Province, People's Republic of China
| |
Collapse
|
143
|
Facciolà A, Venanzi Rullo E, Ceccarelli M, D'Andrea F, Coco M, Micali C, Cacopardo B, Marino A, Cannavò SP, Di Rosa M, Condorelli F, Pellicanò GF, Guarneri C, Nunnari G. Malignant melanoma in HIV: Epidemiology, pathogenesis, and management. Dermatol Ther 2019; 33:e13180. [PMID: 31770477 DOI: 10.1111/dth.13180] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 11/23/2019] [Indexed: 12/23/2022]
Abstract
People affected by immunodeficiency, and especially those infected by HIV, are burdened by a higher risk of developing malignancies. It has been estimated that the incidence of melanoma in HIV-infected people is 2.6-fold higher than in uninfected ones. In this group of patients, melanoma shows a more aggressive phenotype and poorer survival rates compared to HIV-negative people. Standard guidelines of diagnosis and care do not exist yet. Studies suggest high index of suspicion and a low threshold for biopsy in HIV-positive patients regardless of their CD4+ count and the use of standard surgical margins for re-excision procedures. In case of diagnosis of melanoma in HIV-positive patients, a thorough search for metastatic disease is recommended because of the more aggressive course of this cancer in HIV-positive patients. Moreover, to rapidly find out any recurrence or metastatic disease after treatment, these patients need a close follow-up, every 3 months, for the first 2 years and at least twice yearly thereafter. Although surgery remains the main therapeutic option, application of immune checkpoint-based immunotherapy is being studied and seems to be promising. The aim of this review is to present the current knowledge and future options for melanoma diagnosis and treatment in people living with HIV.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Emmanuele Venanzi Rullo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Manuela Ceccarelli
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Flavia D'Andrea
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Mariagiovanna Coco
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Cristina Micali
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Bruno Cacopardo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andrea Marino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Serafinella P Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Fabrizio Condorelli
- Department of Pharmacological Sciences, University of Oriental Piedmont "A. Avogadro", Novara, Italy
| | - Giovanni F Pellicanò
- Department of Human Pathology of the Adult and the Developmental Age "G. Barresi", University of Messina, Messina, Italy
| | - Claudio Guarneri
- Department of Biomedical Sciences and Morphologic and Functional Imaging, University of Messina, Messina, Italy
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
144
|
Abstract
PURPOSE OF REVIEW The last decade witnessed an explosion in immunotherapeutic agent approvals for various malignancies. The success of immune checkpoint inhibitors (CTLA-4 and PD-1/PD-L1) in melanoma quickly sprung to other cancer types and are considered the emerging face of oncology. RECENT FINDINGS Antibodies to CTLA-4 were first to enter the field, quickly followed by PD-1/PD-L1 inhibitors. Combination anti-CTLA4 and anti-PD-1/PD-L1 therapies were investigated, and after demonstrating improved responses, rapidly gained approval. Certain tumor types previously considered non-immunogenic also demonstrated durable responses which has been a remarkable discovery. However, not all tumor types respond to immunotherapies and it is widely recognized that tumor-specific immune inflammatory status predicts the best responders. Ongoing translational work indicates specific upregulation in additional immune checkpoints that circumvent response to anti-CTLA4 and anti-PD-1/PD-L1 antibodies. Here, we provide a comprehensive review of promising therapies on the horizon with unique combinations designed to overcome resistance or expand the pool of treatment responders.
Collapse
|
145
|
Jasim KA, Gesquiere AJ. Ultrastable and Biofunctionalizable Conjugated Polymer Nanoparticles with Encapsulated Iron for Ferroptosis Assisted Chemodynamic Therapy. Mol Pharm 2019; 16:4852-4866. [PMID: 31613630 DOI: 10.1021/acs.molpharmaceut.9b00737] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We report the development of novel tumor-targeted conjugated polymer nanoparticles (CPNPs) carrying iron for chemodynamic therapy (CDT). Tumor cell killing proceeds through ferroptosis, a reactive oxygen species (ROS) mechanism that is not dependent on external activation by, for example, light, as is the case in photodynamic therapy (PDT). The ferroptosis mechanism is also not heavily reliant on oxygen availability and is, therefore, promising for the treatment of hypoxic tumors. In this work, we apply this development to the case study of melanoma, a difficult to treat cancer in advanced stages due to resistance to chemotherapy. The iron-carrying CPNPs reported here are targeted to endothelin-B receptors (EDNRB) through endothelin-3 surface moieties (EDN3-CPNPs). Our results show excellent targeting to tumor cells that overexpress EDNRB, specifically for melanoma and bladder tumor cells. In these cases, efficient cell killing, over 80% at higher doses, was found. Conversely, tumor cells not targeted by the EDN3-CPNPs show little effects of CDT, with tumor cell death under 20% in most cases. The outcomes of our work demonstrate that EDN3-CPNPs enable ferroptosis-assisted CDT and present a new therapeutic avenue for tumor treatment.
Collapse
Affiliation(s)
- Khalaf A Jasim
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States.,NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, United States.,Department of Chemistry, College of Science, Tikrit University, Tikrit 34001, Iraq
| | - Andre J Gesquiere
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States.,NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, United States.,Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida 32816, United States.,The College of Optics and Photonics (CREOL), University of Central Florida, Orlando, Florida 32816, United States
| |
Collapse
|
146
|
Ullrich CI, Aloni R, Saeed MEM, Ullrich W, Efferth T. Comparison between tumors in plants and human beings: Mechanisms of tumor development and therapy with secondary plant metabolites. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:153081. [PMID: 31568956 DOI: 10.1016/j.phymed.2019.153081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 05/24/2023]
Abstract
BACKGROUND Human tumors are still a major threat to human health and plant tumors negatively affect agricultural yields. Both areas of research are developing largely independent of each other. Treatment of both plant and human tumors remains unsatisfactory and novel therapy options are urgently needed. HYPOTHESIS The concept of this paper is to compare cellular and molecular mechanisms of tumor development in plants and human beings and to explore possibilities to develop novel treatment strategies based on bioactive secondary plant metabolites. The interdisciplinary discourse may unravel commonalities and differences in the biology of plant and human tumors as basis for rational drug development. RESULTS Plant tumors and galls develop upon infection by bacteria (e.g. Agrobacterium tumefaciens and A. vitis, which harbor oncogenic T-DNA) and by insects (e.g. gall wasps, aphids). Plant tumors are benign, i.e. they usually do not ultimately kill their host, but they can lead to considerable economic damage due to reduced crop yields of cultivated plants. Human tumors develop by biological carcinogenesis (i.e. viruses and other infectious agents), chemical carcinogenesis (anthropogenic and non-anthropogenic environmental toxic xenobiotics) and physical carcinogenesis (radioactivity, UV-radiation). The majority of human tumors are malignant with lethal outcome. Although treatments for both plant and human tumors are available (antibiotics and apathogenic bacterial strains for plant tumors, cytostatic drugs for human tumors), treatment successes are non-satisfactory, because of drug resistance and the severe adverse side effects. In human beings, attacks by microbes are repelled by cellular immunity (i.e. innate and acquired immune systems). Plants instead display chemical defense mechanisms, whereby constitutively expressed phytoanticipin compounds compare to the innate human immune system, the acquired human immune system compares to phytoalexins, which are induced by appropriate biotic or abiotic stressors. Some chemical weapons of this armory of secondary metabolites are also active against plant galls. There is a mutual co-evolution between plant defense and animals/human beings, which was sometimes referred to as animal plant warfare. As a consequence, hepatic phase I-III metabolization and excretion developed in animals and human beings to detoxify harmful phytochemicals. On the other hand, plants invented "pro-drugs" during evolution, which are activated and toxified in animals by this hepatic biotransformation system. Recent efforts focus on phytochemicals that specifically target tumor-related mechanisms and proteins, e.g. angiogenic or metastatic inhibitors, stimulators of the immune system to improve anti-tumor immunity, specific cell death or cancer stem cell inhibitors, inhibitors of DNA damage and epigenomic deregulation, specific inhibitors of driver genes of carcinogenesis (e.g. oncogenes), inhibitors of multidrug resistance (i.e. ABC transporter efflux inhibitors), secondary metabolites against plant tumors. CONCLUSION The exploitation of bioactive secondary metabolites to treat plant or human tumors bears a tremendous therapeutic potential. Although there are fundamental differences between human and plant tumors, either isolated phytochemicals and their (semi)synthetic derivatives or chemically defined and standardized plant extracts may offer new therapy options to decrease human tumor incidence and mortality as well as to increase agricultural yields by fighting crown galls.
Collapse
Affiliation(s)
- Cornelia I Ullrich
- Department of Biology, Darmstadt University of Technology, Schnittspahnstr. 3-5, Darmstadt 64287, Germany
| | - Roni Aloni
- School of Plant Sciences and Food Security, Tel Aviv University, Tel Aviv 69978, Israel
| | - Mohamed E M Saeed
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz 55128, Germany
| | - Wolfram Ullrich
- Department of Biology, Darmstadt University of Technology, Schnittspahnstr. 3-5, Darmstadt 64287, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz 55128, Germany.
| |
Collapse
|
147
|
Petrelli F, De Stefani A, Trevisan F, Parati C, Inno A, Merelli B, Ghidini M, Bruschieri L, Vitali E, Cabiddu M, Borgonovo K, Ghilardi M, Barni S, Ghidini A. Combination of radiotherapy and immunotherapy for brain metastases: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2019; 144:102830. [PMID: 31733443 DOI: 10.1016/j.critrevonc.2019.102830] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 06/15/2019] [Accepted: 10/26/2019] [Indexed: 01/15/2023] Open
Abstract
Radiotherapy (RT) represents a mainstay in the treatment of brain metastases (BMs) from solid tumors. Immunotherapy (IT) has improved survival of metastatic cancer patients across many tumor types. The combination of RT and IT for the treatment of BMs has a strong rationale, but data on efficacy and safety of this combination is still limited. A systematic search of PubMed, the Cochrane Central Register of Controlled Trials, and EMBASE was conducted. 33 studies were included for a total of 1520 patients, most of them with melanoma (87%). Median pooled OS was 15.9 months (95%CI 13.9-18.1). One- and 2-year OS rates were 55.2% (95% CI 49.3-60.9) and 35.7% (95% CI 30.4-41.3), respectively. Addition of IT to RT was associated with improved OS (HR = 0.54, 95%CI 0.44-0.67; P < 0.001). For patients with BMs from solid tumors, addition of concurrent IT to brain RT is able to increase survival and provide long term control.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy.
| | - Agostina De Stefani
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Francesca Trevisan
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Chiara Parati
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Alessandro Inno
- Oncology Unit, Ospedale Sacro Cuore don Calabria Cancer Care Center, Via Don A. Sempreboni 5, 37024, Negrar, VR, Italy
| | - Barbara Merelli
- Oncology Unit, ASST Papa Giovanni XXIII, Piazza Oms 1, 24127, Bergamo, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Viale F. Sforza 28, 20122, Milano, Italy
| | - Lorenza Bruschieri
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Elisabetta Vitali
- Radiotherapy Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Mary Cabiddu
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Karen Borgonovo
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Mara Ghilardi
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Sandro Barni
- Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | - Antonio Ghidini
- Oncology Unit, Casa di Cura Igea, Via Marcona 69, 20144, Milano, Italy
| |
Collapse
|
148
|
Wu M, Wang Y, Xu Y, Zhu J, Lv C, Sun M, Guo R, Xia Y, Zhang W, Xue C. Indirect comparison between immune checkpoint inhibitors and targeted therapies for the treatment of melanoma. J Cancer 2019; 10:6114-6123. [PMID: 31762821 PMCID: PMC6856565 DOI: 10.7150/jca.32638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
Background: This systematic review and meta-analysis aims to provide comparative and quantitative data about immune checkpoint inhibitor (IMM) and targeted therapy (TAR) in this work. Methods: A literature search was performed with PubMed, Embase, PMC database, and Web of Science databases to identify relevant studies. Hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS), and odds ratios (ORs) for overall response rate (ORR) were estimated. Results: Eighteen manuscripts were ultimately utilized for indirect comparisons. In general, both TAR and IMM can prolong the PFS either by monotherapy, combination therapy with chemotherapy or adjuvant therapy. BRAF inhibitor monotherapy showed superiority over anti-CTLA-4 in OS (HR: 1.28, 95%CI: 0.93-1.75) and best ORR (OR: 12.57, 95%CI: 6.63-23.82), as well as longer PFS (HR: 1.63, 95%CI: 1.00-2.67) and higher best ORR (OR: 3.29, 95%CI: 1.94-5.55) compared with anti-PD-1. However, MEK inhibitor monotherapy showed no priority. When combined with chemotherapy, anti-CTLA-4 showed marginally advantages over MEK inhibitor in OS (HR: 0.68, 95%CI: 0.44-1.03), however no advantage in PFS (HR: 1.12, 95%CI: 0.76-1.64), or ORR (OR: 1.78, 95%CI: 0.70-4.49). For post-operational melanoma patient, adjuvant TAR and adjuvant IMM showed no difference in OS (HR: 1.14, 95%CI: 0.82-1.58) or PFS (HR: 1.20, 95%CI: 0.79-1.83). Moreover, the high-rate adverse events and underlying diseases should be considered during the application of those agents. Conclusions: For the unresectable late-stage melanoma, IMM may be a better choice for the combined treatment with chemotherapy. If the chemotherapy is not tolerable for patients, BRAFi involved TAR can be considered.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yalong Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ji Zhu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chuan Lv
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Mengyan Sun
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Rui Guo
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yu Xia
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Wei Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
149
|
Malissen N, Macagno N, Granjeaud S, Granier C, Moutardier V, Gaudy-Marqueste C, Habel N, Mandavit M, Guillot B, Pasero C, Tartour E, Ballotti R, Grob JJ, Olive D. HVEM has a broader expression than PD-L1 and constitutes a negative prognostic marker and potential treatment target for melanoma. Oncoimmunology 2019; 8:e1665976. [PMID: 31741766 PMCID: PMC6844309 DOI: 10.1080/2162402x.2019.1665976] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 10/27/2022] Open
Abstract
HVEM (Herpes Virus Entry Mediator) engagement of BTLA (B and T Lymphocyte Attenuator) triggers inhibitory signals in T cells and could play a role in evading antitumor immunity. Here, HVEM expression levels in melanoma metastases were analyzed by immunohistochemistry, correlated with overall survival (OS) in 116 patients, and validated by TCGA transcriptomic data. Coincident expression of HVEM and its ligand BTLA was studied in tumor cells and tumor-infiltrating lymphocytes (TILs) by flow cytometry (n = 21) and immunofluorescence (n = 5). Candidate genes controlling HVEM expression in melanoma were defined by bioinformatics studies and validated by siRNA gene silencing. We found that in patients with AJCC stage III and IV melanoma, OS was poorer in those with high HVEM expression on melanoma cells, than in those with a low expression, by immunohistochemistry (p = .0160) or TCGA transcriptomics (p = .0282). We showed a coincident expression of HVEM at the surface of melanoma cells and of BTLA on TILs. HVEM was more widely expressed than PD-L1 in melanoma cells. From a mechanistic perspective, in contrast to PDL1, HVEM expression did not correlate with an IFNγ signature but with an aggressive gene signature. Interestingly, this signature contained MITF, a key player in melanoma biology, whose expression correlated strongly with HVEM. Finally, siRNA gene silencing validated MITF control of HVEM expression. In conclusion, HVEM expression seems to be a prognosis marker and targeting this axis by checkpoint-inhibitors may be of interest in metastatic melanoma.
Collapse
Affiliation(s)
- Nausicaa Malissen
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Nicolas Macagno
- INSERM, MMG, APHM, CHU Timone, Department of Pathology, Aix Marseille University, Marseille, France
| | - Samuel Granjeaud
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| | - Clémence Granier
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Vincent Moutardier
- APHM, CHU Nord, Department of Digestive surgery, Aix Marseille University, Marseille, France
| | - Caroline Gaudy-Marqueste
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Nadia Habel
- INSERM U 1065, Team 1 Nice, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Marion Mandavit
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Bernard Guillot
- Department of Dermatology, CHU Montpellier, Montpellier, France
| | - Christine Pasero
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| | - Eric Tartour
- UMR_S970, HEGP, Centre de recherche cardio-vasculaire, Paris, France
| | - Robert Ballotti
- INSERM U 1065, Team 1 Nice, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Jean-Jacques Grob
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France.,INSERM, CRCM, APHM, CHU Timone, Department of Dermatology and Skin Cancer, Aix Marseille University, Marseille, France
| | - Daniel Olive
- Tumor Immunology Team, IBISA Immunomonitoring platform, Cancer Research Center of Marseille, INSERM U1068, CNRS U7258, Aix-Marseille University, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
150
|
Sinnamon AJ, Gimotty PA, Karakousis GC. Interpreting the Association of First-in-Class Immune Checkpoint Inhibition and Targeted Therapy With Survival in Patients With Stage IV Melanoma-Reply. JAMA Oncol 2019; 4:1136-1137. [PMID: 29902301 DOI: 10.1001/jamaoncol.2018.0898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Andrew J Sinnamon
- Department of Surgery, Oncologic Surgery, Hospital of the University of Pennsylvania, Philadelphia.,Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia
| | - Giorgos C Karakousis
- Department of Surgery, Oncologic Surgery, Hospital of the University of Pennsylvania, Philadelphia
| |
Collapse
|