2351
|
Comito F, Leslie I, Boos L, Furness A, Pickering L, Turajlic S, Larkin J. Oligoprogression After Checkpoint Inhibition in Metastatic Melanoma Treated With Locoregional Therapy: A Single-center Retrospective Analysis. J Immunother 2020; 43:250-255. [PMID: 32796274 DOI: 10.1097/cji.0000000000000333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Checkpoint inhibitors (CPIs) have demonstrated a heterogenous spectrum of response and disease progression that may not be fully captured by conventional response criteria, such as a limited degree of progression, known as oligoprogression, which could benefit from local treatment. We retrospectively analyzed data from all patients diagnosed with metastatic melanoma, who received CPI between January 2006 and March 2018 at Royal Marsden. We enrolled 36 patients who experienced progression in a maximum of 3 metastatic sites, after achieving disease control from therapy with CPI, and were radically treated with the locoregional approach. We carried out Kaplan-Meier analysis to obtain progression free-survival post-first oligoprogression (PFS-PO1), overall survival (OS) post-first oligoprogression, and OS estimates. The median time to oligoprogression from the start of CPI was 12 months. At a median follow-up of 34 months, the median PFS-PO1 was 32 months, with 50% of patients not progressed at the time of the data cutoff. The median OS-post-first oligoprogression was not reached. At a median follow-up of 52 months (from the first cycle of CPI), the median OS was not reached, with 75% of patients alive at the time of analysis. Univariate and multivariate analyses demonstrated that baseline American Joint Committee on Cancer stage IV M1a or M1b is associated with a longer PFS-PO1 compared with stage M1c or M1d. We observed that local therapy for oligoprogression after CPI can result in durable disease control, suggesting that locoregional treatment should be considered in patients being treated with immunotherapy. However, prospective evaluation, perhaps in randomized trials, is needed.
Collapse
Affiliation(s)
| | | | | | | | | | - Samra Turajlic
- The Royal Marsden NHS Foundation Trust
- Translational Cancer Therapeutics Laboratory, the Francis Crick Institute, London, UK
| | | |
Collapse
|
2352
|
Schoenfeld JD, Hanna GJ, Jo VY, Rawal B, Chen YH, Catalano PS, Lako A, Ciantra Z, Weirather JL, Criscitiello S, Luoma A, Chau N, Lorch J, Kass JI, Annino D, Goguen L, Desai A, Ross B, Shah HJ, Jacene HA, Margalit DN, Tishler RB, Wucherpfennig KW, Rodig SJ, Uppaluri R, Haddad RI. Neoadjuvant Nivolumab or Nivolumab Plus Ipilimumab in Untreated Oral Cavity Squamous Cell Carcinoma: A Phase 2 Open-Label Randomized Clinical Trial. JAMA Oncol 2020; 6:1563-1570. [PMID: 32852531 PMCID: PMC7453348 DOI: 10.1001/jamaoncol.2020.2955] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022]
Abstract
Importance Novel approaches are needed to improve outcomes in patients with squamous cell carcinoma of the oral cavity. Neoadjuvant immunotherapy given prior to surgery and combining programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immune checkpoint inhibitors are 2 strategies to enhance antitumor immune responses that could be of benefit. Design, Setting, and Participants In this randomized phase 2 clinical trial conducted at 1 academic center, 29 patients with untreated squamous cell carcinoma of the oral cavity (≥T2, or clinically node positive) were enrolled between 2016 to 2019. Interventions Treatment was administered with nivolumab, 3 mg/kg, weeks 1 and 3, or nivolumab and ipilimumab (ipilimumab, 1 mg/kg, given week 1 only). Patients had surgery 3 to 7 days following cycle 2. Main Outcomes and Measures Safety and volumetric response determined using bidirectional measurements. Secondary end points included pathologic and objective response, progression-free survival (PFS), and overall survival. Multiplex immunofluorescence was used to evaluate primary tumor immune markers. Results Fourteen patients were randomized to nivolumab (N) and 15 patients to nivolumab/ipilimumab (N+I) (mean [SD] age, 62 [12] years; 18 men [62%] and 11 women [38%]). The most common subsite was oral tongue (n = 16). Baseline clinical staging included patients with T2 (n = 20) or greater (n = 9) T stage and 17 patients (59%) with node-positive disease. Median time from cycle 1 to surgery was 19 days (range, 7-21 days); there were no surgical delays. There were toxic effects at least possibly related to study treatment in 21 patients, including grade 3 to 4 events in 2 (N), and 5 (N+I) patients. One patient died of conditions thought unrelated to study treatment (postoperative flap failure, stroke). There was evidence of response in both the N and N+I arms (volumetric response 50%, 53%; pathologic downstaging 53%, 69%; RECIST response 13%, 38%; and pathologic response 54%, 73%, respectively). Four patients had major/complete pathologic response greater than 90% (N, n = 1; N+I, n = 3). With 14.2 months median follow-up, 1-year progression-free survival was 85% and overall survival was 89%. Conclusions and Relevance Treatment with N and N+I was feasible prior to surgical resection. We observed promising rates of response in both arms, supporting further neoadjuvant studies with these agents. Trial Registration ClinicalTrials.gov Identifier: NCT02919683.
Collapse
Affiliation(s)
- Jonathan D. Schoenfeld
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Glenn J. Hanna
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Vickie Y. Jo
- Brigham and Women's Hospital, Boston, Massachusetts
| | - Bhupendra Rawal
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- PRA Health Sciences, Boston, Massachusetts
| | - Yu-Hui Chen
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Paul S. Catalano
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Ana Lako
- Brigham and Women's Hospital, Boston, Massachusetts
- Bristol-Myers Squibb, Boston, Massachusetts
| | - Zoe Ciantra
- Brigham and Women's Hospital, Boston, Massachusetts
| | - Jason L. Weirather
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Shana Criscitiello
- Brigham and Women's Hospital, Boston, Massachusetts
- Beth-Israel Deaconess Medical Center, Boston, Massachusetts
| | - Adrienne Luoma
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Nicole Chau
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- British Columbia Cancer, Vancouver, Canada
| | - Jochen Lorch
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jason I. Kass
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Donald Annino
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Laura Goguen
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Anupam Desai
- Beth-Israel Deaconess Medical Center, Boston, Massachusetts
| | - Brendan Ross
- Brigham and Women's Hospital, Boston, Massachusetts
- McGill Medical School, Montreal, Canada
| | - Hina J. Shah
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Heather A. Jacene
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Danielle N. Margalit
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Roy B. Tishler
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | | - Scott J. Rodig
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Ravindra Uppaluri
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Robert I. Haddad
- Brigham and Women's Hospital, Boston, Massachusetts
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2353
|
Winge-Main AK, Wälchli S, Inderberg EM. T cell receptor therapy against melanoma-Immunotherapy for the future? Scand J Immunol 2020; 92:e12927. [PMID: 32640053 DOI: 10.1111/sji.12927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
Malignant melanoma has seen monumental changes in treatment options the last decade from the very poor results of dacarbazine treatment to the modern-day use of targeted therapies and immune checkpoint inhibitors. Melanoma has a high mutational burden making it more capable of evoking immune responses than many other tumours. Even when considering double immune checkpoint blockade with anti-CTLA-4 and anti-PD-1, we still have far to go in melanoma treatment as 50% of patients with metastatic disease do not respond to current treatment. Alternative immunotherapy should therefore be considered. Since melanoma has a high mutational burden, it is considered more immunogenic than many other tumours. T cell receptor (TCR) therapy could be a possible way forward, either alone or in combination, to improve the response rates of this deadly disease. Melanoma is one of the cancers where TCR therapy has been frequently applied. However, the number of antigens targeted remains fairly limited, although advanced personalized therapies aim at also targeting private mutations. In this review, we look at possible aspects of targeting TCR therapy towards melanoma and provide an implication of its use in the future.
Collapse
Affiliation(s)
- Anna K Winge-Main
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2354
|
Johnson DB, Jakubovic BD, Sibaud V, Sise ME. Balancing Cancer Immunotherapy Efficacy and Toxicity. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2020; 8:2898-2906. [PMID: 32599218 PMCID: PMC7318967 DOI: 10.1016/j.jaip.2020.06.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022]
Abstract
Anti-programmed cell death-1 receptor/programmed cell death-1 receptor ligand-directed therapies are transforming cancer care, with durable antitumor responses observed in multiple cancer types. Toxicities arising from therapy are autoimmune in nature and may affect essentially any organ system. The immunologic basis of such toxities is complex, with contributions from T-cell activation and autoantibody generation. Although less recognized, hypersensitivity reactions are also possible. Although most toxicities resolve with systemic corticosteroids, some require second-line immunosuppression. Furthermore, the safety of drug rechallenge is not well characterized, with variable rates of toxicity flares arising with re-exposure. Herein, we review toxicities of immune checkpoint inhibitor therapies, particularly focusing on issues that allergists/immunologists may clinically encounter, including interstitial nephritis, skin toxicity, and risks associated with immunotherapy rechallenge.
Collapse
Affiliation(s)
- Douglas B Johnson
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn.
| | - Baruch D Jakubovic
- Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Vincent Sibaud
- Department of Oncodermatology, Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| | - Meghan E Sise
- Renal Division, Department of Internal Medicine, Massachusetts General Hospital, Boston, Mass
| |
Collapse
|
2355
|
Tang X, Zhang H, Cui Y, Wang L, Wang Z, Zhang Y, Huo J, Cai J, Rinaldi G, Bhagavathula AS, Xiaopeng Y. Postmenopausal exogenous hormone therapy and Melanoma risk in women: A systematic review and time-response meta-analysis. Pharmacol Res 2020; 160:105182. [PMID: 32890740 DOI: 10.1016/j.phrs.2020.105182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 01/11/2023]
Abstract
A favourable option to management symptoms during menopausal transition is menopausal hormone therapy (MHT) but relation among MHT and risk of melanoma is controversial. This study aims to identify, analyse and present the evidence surrounding post-menopausal exogenous hormone therapy and the risk for melanoma in women. A systematic searches of database was conducted in PubMed/MEDLINE, Scopus, and Cochrane without time, region, and language restrictions from inception to April 2020. The DerSimonian and Laird random-effects model was used to estimate combined risk ratio (RR) and 95% confidence intervals (CI). Subgroup analysis and time-response analysis was conducted based on the formulation of used hormone and length of hormone therapy. Combined results from fourteen studies that containing 19 arms with 1,164,077 participants which 4273 of them had melanoma cancer showed increase risk of melanoma in the hormone-treated versus control group 1.14 (95% CI 1.05-1.24, I2: 21%). The stronger and significant relationship between MHT and risk of melanoma was in participants who used oestrogen formulation (RR 1.32, 95% CI 1.17-1.49, I2 = 0%). Moreover, a significant non-linear time-response relation between MHT and melanoma was also in initial three years of MHT (Coef1 = 0.2423, p1 < 0.01). This study reveals a significant direct relationship between the MHT and risk of melanoma in postmenopausal women.
Collapse
Affiliation(s)
- Xiaoling Tang
- Malignant Tumor TCM "Yi Qi Qing Du" Key Research Office, Jiangxi Academy of Traditional Chinese Medicine, Nangchang, Jiangxi 330046, China
| | - Hongcan Zhang
- Outpatient Clinic of Ethnomedicine, Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, China
| | - Ying Cui
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Liqin Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanchang University, Nangchang, Jiangxi 330006, China
| | - Zhu Wang
- Malignant Tumor TCM "Yi Qi Qing Du" Key Research Office, Jiangxi Academy of Traditional Chinese Medicine, Nangchang, Jiangxi 330046, China
| | - Yajing Zhang
- Institute of Literature and Information, Jiangxi Academy of Traditional Chinese Medicine, Nangchang, Jiangxi 330046, China
| | - Jinzhi Huo
- Obstetrics and Gynecology Clinic, No. 963 Hospital, Jiamusi City, Heilongjiang Province 154000, China
| | - Jinfeng Cai
- Department of Reproductive Center, Kunming City Maternal and Child Health Hospital, Kunming, Yunnan 650031, China
| | - Giulia Rinaldi
- Guys and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Ying Xiaopeng
- Department of Reproductive Center, Kunming City Maternal and Child Health Hospital, Kunming, Yunnan 650031, China.
| |
Collapse
|
2356
|
Lee AY, Brady MS. Neoadjuvant immunotherapy for melanoma. J Surg Oncol 2020; 123:782-788. [PMID: 33002195 DOI: 10.1002/jso.26229] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Clinical trials have demonstrated the efficacy of immunotherapy, especially checkpoint blockade inhibitors, in the treatment of patients with metastatic melanoma. More recently, improvements in survival have been reported in patients with high-risk resectable melanoma when these agents are used in the adjuvant setting. Increasing interest in neoadjuvant immunotherapy for high-risk resectable melanoma has been fueled by early reports of significant efficacy. We review the rationale and data behind utilizing neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Ann Y Lee
- Department of Surgery, NYU Langone Health, New York, New York, USA
| | - Mary S Brady
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
2357
|
Michielin O, Atkins MB, Koon HB, Dummer R, Ascierto PA. Evolving impact of long-term survival results on metastatic melanoma treatment. J Immunother Cancer 2020; 8:e000948. [PMID: 33037115 PMCID: PMC7549477 DOI: 10.1136/jitc-2020-000948] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Melanoma treatment has been revolutionized over the past decade. Long-term results with immuno-oncology (I-O) agents and targeted therapies are providing evidence of durable survival for a substantial number of patients. These results have prompted consideration of how best to define long-term benefit and cure. Now more than ever, oncologists should be aware of the long-term outcomes demonstrated with these newer agents and their relevance to treatment decision-making. As the first tumor type for which I-O agents were approved, melanoma has served as a model for other diseases. Accordingly, discussions regarding the value and impact of long-term survival data in patients with melanoma may be relevant in the future to other tumor types. Current findings indicate that, depending on the treatment, over 50% of patients with melanoma may gain durable survival benefit. The best survival outcomes are generally observed in patients with favorable prognostic factors, particularly normal baseline lactate dehydrogenase and/or a low volume of disease. Survival curves from melanoma clinical studies show a plateau at 3 to 4 years, suggesting that patients who are alive at the 3-year landmark (especially in cases in which treatment had been stopped) will likely experience prolonged cancer remission. Quality-of-life and mixture-cure modeling data, as well as metrics such as treatment-free survival, are helping to define the value of this long-term survival. In this review, we describe the current treatment landscape for melanoma and discuss the long-term survival data with immunotherapies and targeted therapies, discussing how to best evaluate the value of long-term survival. We propose that some patients might be considered functionally cured if they have responded to treatment and remained treatment-free for at least 2 years without disease progression. Finally, we consider that, while there have been major advances in the treatment of melanoma in the past decade, there remains a need to improve outcomes for the patients with melanoma who do not experience durable survival.
Collapse
Affiliation(s)
- Olivier Michielin
- Oncology Department, Precision Oncology Center, Lausanne, Switzerland
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Michael B Atkins
- Medical Oncology, Georgetown Lombardi Comprehensive Cancer Center and Oncology Academic Department, Georgetown University Medical Center, Washington, DC, USA
| | - Henry B Koon
- Clinical Research, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Paolo Antonio Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| |
Collapse
|
2358
|
Han S, Shuen WH, Wang WW, Nazim E, Toh HC. Tailoring precision immunotherapy: coming to a clinic soon? ESMO Open 2020; 5 Suppl 1:e000631. [PMID: 33558033 PMCID: PMC7046383 DOI: 10.1136/esmoopen-2019-000631] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/29/2019] [Accepted: 02/04/2020] [Indexed: 12/23/2022] Open
Abstract
Cell-based and antibody-based cancer immunotherapies have been widely tested across increasing numbers of cancers with an unprecedented number of successful practice-changing immunotherapy clinical trials, achieving significant survival outcomes and, characteristically, some very long-term survivors. Still, a sizeable proportion of patients, especially with solid tumours, do not benefit from immunotherapy. Here, we summarise key literature on immunotherapy biomarkers and resistance mechanisms and discuss potential strategies to overcome such resistance to improve patient outcomes. The ever-expanding understanding of the tumour-immune interaction and the tumour microenvironment allows a real opportunity to identify predictive biomarkers and tailor immune-based therapies, including designing rational combination drugs to enhance clinical outcomes, and to identify patients most likely to benefit from immunotherapy. Where there has never been a precision chemotherapy clinic in the last 70 years since its inception, even with no shortage of trying, the hope and evolution of a functional precision immunotherapy cancer clinic is a much more likely reality.
Collapse
Affiliation(s)
- Shuting Han
- Division of Medical Ocology, National Cancer Centre Singapore, Singapore, Singapore
| | - Wai Ho Shuen
- Division of Medical Ocology, National Cancer Centre Singapore, Singapore, Singapore
| | - Who-Whong Wang
- Division of Medical Ocology, National Cancer Centre Singapore, Singapore, Singapore
| | - Esdy Nazim
- Division of Medical Ocology, National Cancer Centre Singapore, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Ocology, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
2359
|
Robert C, Long GV, Brady B, Dutriaux C, Di Giacomo AM, Mortier L, Rutkowski P, Hassel JC, McNeil CM, Kalinka EA, Lebbé C, Charles J, Hernberg MM, Savage KJ, Chiarion-Sileni V, Mihalcioiu C, Mauch C, Arance A, Cognetti F, Ny L, Schmidt H, Schadendorf D, Gogas H, Zoco J, Re S, Ascierto PA, Atkinson V. Five-Year Outcomes With Nivolumab in Patients With Wild-Type BRAF Advanced Melanoma. J Clin Oncol 2020; 38:3937-3946. [PMID: 32997575 PMCID: PMC7676881 DOI: 10.1200/jco.20.00995] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The CheckMate 066 trial investigated nivolumab monotherapy as first-line treatment for patients with previously untreated BRAF wild-type advanced melanoma. Five-year results are presented herein.
Collapse
Affiliation(s)
- Caroline Robert
- Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | | | - Caroline Dutriaux
- Hôpital Saint André Centre Hospitalier Universitaire, Bordeaux, France
| | | | - Laurent Mortier
- University of Lille, INSERM U1189, Service de Dermatologie, Chu Lille, Lille, France
| | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Jessica C Hassel
- National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Catriona M McNeil
- Chris O'Brien Lifehouse, Melanoma Institute Australia, and Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Ewa Anna Kalinka
- Polish Mother's Memorial Hospital-Research Institute, Lodz, Poland
| | - Céleste Lebbé
- Université de Paris, INSERM U976, and Dermatology and CIC, AP-HP, Saint Louis Hospital, Paris, France
| | - Julie Charles
- Grenoble Alpes University Hospital, INSERM U1209, Grenoble Alpes University, Grenoble, France
| | | | - Kerry J Savage
- British Columbia Cancer Agency, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vanna Chiarion-Sileni
- Oncology Institute of Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | | | - Cornelia Mauch
- University Hospital Cologne and Centrum für Integrierte Onkologie Köln, Bonn, Germany
| | - Ana Arance
- Hospital Clínic Barcelona, Barcelona, Spain
| | | | - Lars Ny
- Department of Oncology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Dirk Schadendorf
- Department of Dermatology, Comprehensive Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium, Heidelberg, Germany
| | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Victoria Atkinson
- Princess Alexandra Hospital, Woolloongabba, and Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
| |
Collapse
|
2360
|
Gamat-Huber M, Jeon D, Johnson LE, Moseman JE, Muralidhar A, Potluri HK, Rastogi I, Wargowski E, Zahm CD, McNeel DG. Treatment Combinations with DNA Vaccines for the Treatment of Metastatic Castration-Resistant Prostate Cancer (mCRPC). Cancers (Basel) 2020; 12:cancers12102831. [PMID: 33008010 PMCID: PMC7601088 DOI: 10.3390/cancers12102831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
Simple Summary The only vaccine approved by FDA as a treatment for cancer is sipuleucel-T, a therapy for patients with metastatic castration-resistant prostate cancer (mCRPC). Most investigators studying anti-tumor vaccines believe they will be most effective as parts of combination therapies, rather than used alone. Unfortunately, the cost and complexity of sipuleucel-T makes it difficult to feasibly be used in combination with many other agents. In this review article we discuss the use of DNA vaccines as a simpler vaccine approach that has demonstrated efficacy in several animal species. We discuss the use of DNA vaccines in combination with traditional treatments for mCRPC, and other immune-modulating treatments, in preclinical and early clinical trials for patients with mCRPC. Abstract Metastatic castration-resistant prostate cancer (mCRPC) is a challenging disease to treat, with poor outcomes for patients. One antitumor vaccine, sipuleucel-T, has been approved as a treatment for mCRPC. DNA vaccines are another form of immunotherapy under investigation. DNA immunizations elicit antigen-specific T cells that cause tumor cell lysis, which should translate to meaningful clinical responses. They are easily amenable to design alterations, scalable for large-scale manufacturing, and thermo-stable for easy transport and distribution. Hence, they offer advantages over other vaccine formulations. However, clinical trials with DNA vaccines as a monotherapy have shown only modest clinical effects against tumors. Standard therapies for CRPC including androgen-targeted therapies, radiation therapy and chemotherapy all have immunomodulatory effects, which combined with immunotherapies such as DNA vaccines, could potentially improve treatment. In addition, many investigational drugs are being developed which can augment antitumor immunity, and together with DNA vaccines can further enhance antitumor responses in preclinical models. We reviewed the literature available prior to July 2020 exploring the use of DNA vaccines in the treatment of prostate cancer. We also examined various approved and experimental therapies that could be combined with DNA vaccines to potentially improve their antitumor efficacy as treatments for mCRPC.
Collapse
|
2361
|
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
|
2362
|
Austin M, Kluger H. Targeting Innate Immunity to Treat Cancer. Cancers (Basel) 2020; 12:cancers12102723. [PMID: 32977403 PMCID: PMC7597964 DOI: 10.3390/cancers12102723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 11/22/2022] Open
|
2363
|
Abstract
PD-1 and CTLA-4 are checkpoint inhibitors of the immune response in cancer, making them the target molecules for the development of therapeutic antibodies. US2019161548 patent describes a bispecific antibody capable of specifically binding to PD-1 and CTLA-4 that induced the proliferation and activation of CD8+ cells, as well as the expression of induclble co-stimulator in CD4+ T cells. Clinical trials to evaluate safety, dose-limiting toxicities and maximum tolerated/administered dose are still in the patient recruitment phase, but it will be of great interest to the scientific and medical community to know if the first bispecific anti-PD-1/CLTA-4 antibody, exceeds expectations and exceeds action of the combination of nivolumab and epilimumab in the treatment of cancer.
Collapse
|
2364
|
Abstract
PURPOSE OF REVIEW The aim of the review is to describe recent advances in our understanding of how multiple myeloma interacts with its cellular and molecular neighbours in the bone marrow microenvironment, and how this may provide targets for prognostication and prevention. RECENT FINDINGS The bone marrow microenvironment in myeloma is beginning to yield targets that are amenable to therapy. A number of trials demonstrate some clinical efficacy in heavily pretreated disease. The challenge remains for how and when these therapeutic interventions are of particular benefit early in disease progression. SUMMARY Multiple myeloma is rarely curable and its interactions with the bone marrow microenvironment are evident. However, separating cause from effect remains a challenge. We propose that targeting specific niches within the bone marrow will yield therapies that have the potential for significant benefit in myeloma and may facilitate earlier intervention to disrupt an environment that is permissive for myeloma progression.
Collapse
|
2365
|
Kang K, Xie F, Mao J, Bai Y, Wang X. Significance of Tumor Mutation Burden in Immune Infiltration and Prognosis in Cutaneous Melanoma. Front Oncol 2020; 10:573141. [PMID: 33072607 PMCID: PMC7531222 DOI: 10.3389/fonc.2020.573141] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Melanoma is highly immunogenic and therefore suitable for immunotherapy, but the efficacy is limited by response rate. In several types of tumor, tumor mutation burden (TMB) and immune infiltration have been reported to predict the response to immunotherapy, although each has its limitations. In the current study, we aimed to explore the association of TMB with immune infiltration and prognosis in cutaneous melanoma. Methods: The data of cutaneous melanoma used for analyses was downloaded from The Cancer Genome Atlas (TCGA) database. The mutation data was sorted using "maftools" R package. TMB was estimated and then patients were divided into two groups based on TMB. The association of TMB with prognosis and clinical characteristics was explored. Differential analysis between two TMB groups was performed using "DESeq2" R package to identify differentially expressed genes (DEGs). The function enrichment analyses of DEGs were conducted to screen critical pathways. Besides, DEGs were further filtered to identify two hub genes, based on which a risk score model and nomogram for predicting prognosis were conducted, and the validation was performed using three datasets from Gene Expression Omnibus (GEO) database. Finally, CIBERSORT algorithm and TIMER database were used to assess the effect of TMB and hub genes on immune infiltration. Results: The most common mutation was C > T, and the top three frequently mutated genes were TTN, MUC16, and BRAF. Higher TMB indicated better survival outcomes and lower pathological stages. 735 DEGs were identified and mainly involved in immune-related and adhesion-related pathways. The risk score model and nomogram were validated using receiver operating characteristic (ROC) curves and calibration curves, and exhibited relatively high predictive capability. Decision curve analysis (DCA) was used to assess clinical benefit. As for immune infiltration, the proportion was higher for macrophages M1 and M2 in the high-TMB group, while lower for memory B cells and regulatory T cells. Conclusions: In cutaneous melanoma, TMB was positively correlated with prognosis. The risk score model and nomogram can be conveniently used to predict prognosis. The association of TMB with immune infiltration can help improve the predicting methods for the response to immunotherapy.
Collapse
Affiliation(s)
- Kai Kang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fucun Xie
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinzhu Mao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Department of Hepatobiliary Surgery, First Central Hospital, Tianjin, China
| | - Xiang Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2366
|
Criscitiello C, Marra A, Morganti S, Zagami P, Viale G, Esposito A, Curigliano G. Pretreatment Blood Parameters Predict Efficacy from Immunotherapy Agents in Early Phase Clinical Trials. Oncologist 2020; 25:e1732-e1742. [PMID: 32785940 DOI: 10.1634/theoncologist.2020-0518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Peripheral blood parameters are correlated to immune-checkpoint inhibitor efficacy in solid tumors, such as melanoma and non-small cell lung cancer. Few data are currently available on the prognostic role of these immune-inflammatory biomarkers for other solid tumors and immunotherapy combinations. MATERIAL AND METHODS From August 2014 to May 2019, 153 patients with metastatic solid tumors were enrolled in phase I clinical trials testing immunotherapy both as single agents and as combinations. Primary endpoint was to evaluate the impact of baseline blood parameters on progression-free survival (PFS) and overall survival (OS). RESULTS The most common tumor types were gastrointestinal, breast, and gynecological cancers (22.9%, 22.2%, and 15.0%, respectively). Higher lactate dehydrogenase (LDH) and derived neutrophil-to-lymphocyte ratio (dNLR) were independently associated with reduced PFS (hazard ratio [HR], 1.97; 95% confidence interval [CI], 1.30-2.99; p = .001, and HR, 2.29; 95% CI, 1.39-3.77; p = .001, respectively) and reduced OS (HR, 2.04; 95% CI, 1.26-3.28; p = .004, and HR, 2.06; 95% CI, 1.12-3.79; p = .02, respectively). In the subgroup analysis, (single agent vs. combination), patients at "good" (dNLR <3 and LDH < upper limit of normal [ULN]) and "intermediate and poor" (dNLR >3 and/or LDH > ULN) risk had higher and lower PFS, respectively (p for interaction = .002). Conversely, patients receiving monotherapy presented statistically significant difference in OS according to the risk group, whereas this effect was not observed for those treated with combinations (p for interaction = .004). CONCLUSION Elevated LDH and dNLR are associated with poorer survival outcomes in patients treated with immunotherapy in phase I clinical trials, regardless of tumor type. These parameters represent an easy tool that might be considered as stratification factors in immunotherapy-based clinical trials. IMPLICATIONS FOR PRACTICE In this retrospective cohort study of 153 patients with metastatic solid tumors treated with immunotherapy in the context of phase I clinical trials, elevated baseline lactate dehydrogenase and derived neutrophil-to-lymphocyte ratio were associated with reduced survival regardless of tumor subtype. If prospectively validated, these parameters might represent low-cost and easy biomarkers that could help patient selection for early phase immunotherapy trials and be applied as a stratification factor in randomized studies testing immunotherapy agents.
Collapse
Affiliation(s)
- Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonio Marra
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Paola Zagami
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
2367
|
Champiat S, Tselikas L, Farhane S, Raoult T, Texier M, Lanoy E, Massard C, Robert C, Ammari S, De Baère T, Marabelle A. Intratumoral Immunotherapy: From Trial Design to Clinical Practice. Clin Cancer Res 2020; 27:665-679. [PMID: 32943460 DOI: 10.1158/1078-0432.ccr-20-0473] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/10/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
Systemic immunotherapies such as immune checkpoint blockade targeted at PD(L)1 and CTLA4 have demonstrated their ability to provide durable tumor responses and long-term overall survival benefits for some patients in several solid tumor types. However, a majority of patients remain resistant to these treatments and a significant proportion of them develop severe autoimmune and inflammatory adverse events. Preclinical studies have demonstrated that intratumoral injections of immunostimulatory products (oncolytics, pattern recognition receptor agonists,…) that are able to trigger type I IFN release and enhance tumor antigen presentation on immune cells could generate a strong antitumor immunity and overcome the resistance to systemic immune checkpoint blockade therapies. The intratumoral immunotherapy strategies that are currently in clinical development offer a unique therapeutic and exploratory setting to better understand the immune contexture across tumor lesions of patients with metastatic cancer. Also these local therapeutic products could turn cold tumors into hot and improve the response rates to cancer immunotherapies while diminishing their systemic exposure and toxicities. Intratumoral immunotherapies could prime or boost the immunity against tumors and therefore radically change the combinatorial therapeutic strategies currently pursued for metastatic and local cancers to improve their long-term survival. We aimed to review and discuss the scientific rationale for intratumoral immunotherapy, the challenges raised by this strategy in terms of drug development within clinical trials and the current state-of-the-art regarding the clinical practice of this innovative approach.
Collapse
Affiliation(s)
- Stéphane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Siham Farhane
- Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Matthieu Texier
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Emilie Lanoy
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Département de Médecine Oncologique (DMO), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Samy Ammari
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry De Baère
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
2368
|
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
|
2369
|
Aiken TJ, Stahl CC, Schwartz PB, Barrett J, Acher AW, Lemaster D, Leverson G, Weber S, Neuman H, Abbott DE. Sentinel lymph node biopsy is associated with increased cost in higher risk thin melanoma. J Surg Oncol 2020; 123:104-109. [PMID: 32939750 DOI: 10.1002/jso.26225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 02/02/2023]
Abstract
INTRODUCTION National Comprehensive Cancer Network guidelines recommend that sentinel lymph node biopsy (SLNB) be discussed with patients with thin melanoma at higher risk for lymph node metastasis (T1b or T1a with positive deep margins, lymphovascular invasion, or high mitotic index). We examined the association between SLNB and resource utilization in this cohort. METHODS We conducted a retrospective cohort study of patients that underwent wide local excision for higher risk thin melanomas from 2009 to 2018 at a tertiary care center. Patients who underwent SLNB were compared to those who did not undergo SLNB with regard to resource utilization, including total hospital cost. RESULTS A total of 70 patients were included in the analysis and 50 patients (71.4%) underwent SLNB. SLNB was associated with increased hospital costs ($6700 vs. $3767; p < .01) and increased operative time (68.5 vs. 36.0 min; p < .01). This cost difference persisted in multivariable regression (p < .01). Of patients who underwent successful SLN mapping, 3 out of 49 patients had a positive SLN (6.1%). The cost to identify a single positive sentinel lymph node (SLN) was $47,906. CONCLUSION In patients with a higher risk of thin melanoma, SLNB is associated with increased cost despite a low likelihood of SLN positivity. These data better inform patient-provider discussions as the role of SLNB in thin melanoma evolves.
Collapse
Affiliation(s)
- Taylor J Aiken
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Christopher C Stahl
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Patrick B Schwartz
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - James Barrett
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Alexandra W Acher
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Deborah Lemaster
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Glen Leverson
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Sharon Weber
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Heather Neuman
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| | - Daniel E Abbott
- Department of Surgery, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, USA
| |
Collapse
|
2370
|
Gaissmaier L, Elshiaty M, Christopoulos P. Breaking Bottlenecks for the TCR Therapy of Cancer. Cells 2020; 9:E2095. [PMID: 32937956 PMCID: PMC7564186 DOI: 10.3390/cells9092095] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors have redefined the treatment of cancer, but their efficacy depends critically on the presence of sufficient tumor-specific lymphocytes, and cellular immunotherapies develop rapidly to fill this gap. The paucity of suitable extracellular and tumor-associated antigens in solid cancers necessitates the use of neoantigen-directed T-cell-receptor (TCR)-engineered cells, while prevention of tumor evasion requires combined targeting of multiple neoepitopes. These can be currently identified within 2 weeks by combining cutting-edge next-generation sequencing with bioinformatic pipelines and used to select tumor-reactive TCRs in a high-throughput manner for expeditious scalable non-viral gene editing of autologous or allogeneic lymphocytes. "Young" cells with a naive, memory stem or central memory phenotype can be additionally armored with "next-generation" features against exhaustion and the immunosuppressive tumor microenvironment, where they wander after reinfusion to attack heavily pretreated and hitherto hopeless neoplasms. Facilitated by major technological breakthroughs in critical manufacturing steps, based on a solid preclinical rationale, and backed by rapidly accumulating evidence, TCR therapies break one bottleneck after the other and hold the promise to become the next immuno-oncological revolution.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Mariam Elshiaty
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| |
Collapse
|
2371
|
de Miguel M, Calvo E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell 2020; 38:326-333. [PMID: 32750319 DOI: 10.1016/j.ccell.2020.07.004] [Citation(s) in RCA: 324] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/18/2020] [Accepted: 07/10/2020] [Indexed: 02/03/2023]
Abstract
Even though the immuno-oncology (IO) era has achieved many successes, some signs of research development deceleration are arising. Recently, the number of FDA immunotherapy approvals has decreased concurrently with a decline in the relative number of patients recruited to these trials. Identifying the unique features of IO treatments and taking them into consideration on clinical research will lead to a better evaluation of these agents and patient outcomes. In this review, we discuss current challenges and new potential approaches to implement rationally designed clinical trials of IO drugs, particularly those targeting immune checkpoints.
Collapse
Affiliation(s)
- Maria de Miguel
- START Madrid-HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Calle Oña, 10, 28050 Madrid, Spain
| | - Emiliano Calvo
- START Madrid-HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Calle Oña, 10, 28050 Madrid, Spain.
| |
Collapse
|
2372
|
Lynch FA, Katona L, Jefford M, Smith AB, Shaw J, Dhillon HM, Ellen S, Phipps-Nelson J, Lai-Kwon J, Milne D, Russell L, Dax V, Diggens J, Kent H, Button-Sloan A, Elliott J, Shackleton M, Burridge H, Ftanou M. Feasibility and Acceptability of Fear-Less: A Stepped-Care Program to Manage Fear of Cancer Recurrence in People with Metastatic Melanoma. J Clin Med 2020; 9:jcm9092969. [PMID: 32937942 PMCID: PMC7565154 DOI: 10.3390/jcm9092969] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 12/27/2022] Open
Abstract
Immunotherapies and targeted therapies have revolutionised treatment of metastatic melanoma and improved survival rates. However, survivors treated with novel therapies are vulnerable to high levels of fear of cancer recurrence or progression (FCR). Existing FCR interventions have rarely been trialled in people with advanced cancer. The current study aimed to evaluate the acceptability and feasibility of Fear-Less: a stepped-care model to treat FCR in people with metastatic melanoma treated with immunotherapy or targeted therapy. Sixty-one outpatients with metastatic melanoma were screened using the Fear of Cancer Recurrence Inventory Short Form (FCRI-SF) and Fear of Progression Questionnaire Short Form (FoP-Q-SF). Survivors with subthreshold FCR were stratified to a self-management intervention while those with clinical levels of FCR were provided with an individual therapy, Conquer Fear. Survivor experience surveys and rescreening were administered post-intervention completion. Results indicated that Fear-Less was an acceptable and feasible FCR intervention. Results provided preliminary support for the potential impact of Fear-Less in reducing FCR. Fear-Less is a promising first step in providing an acceptable and feasible stepped-care model to treat FCR in survivors with metastatic disease.
Collapse
Affiliation(s)
- Fiona A Lynch
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
- Correspondence: ; Tel.: +61-3-8559-8236
| | - Lynda Katona
- Department of Psychology and Consultation Liaison Psychiatry, Alfred Health, Melbourne, VIC 3004, Australia; (L.K.); (H.K.)
| | - Michael Jefford
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (M.J.); (J.L.-K.)
- Australian Cancer Survivorship Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Allan Ben Smith
- Centre for Oncology Education and Research Translation, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Kensington, NSW 2052, Australia;
| | - Joanne Shaw
- Psycho-Oncology Co-operative Research Group (PoCoG), School of Psychology, The University of Sydney, Sydney, NSW 2006, Australia; (J.S.); (H.M.D.)
| | - Haryana M Dhillon
- Psycho-Oncology Co-operative Research Group (PoCoG), School of Psychology, The University of Sydney, Sydney, NSW 2006, Australia; (J.S.); (H.M.D.)
- Centre for Medical Psychology & Evidence-based Decision-making, School of Psychology, The University of Sydney, Sydney, NSW 2006, Australia
| | - Steve Ellen
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
- Department of Psychiatry, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jo Phipps-Nelson
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia;
- Health Services and Implementation Science, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
| | - Julia Lai-Kwon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (M.J.); (J.L.-K.)
| | - Donna Milne
- Health Services and Implementation Science, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Department of Skin and Melanoma Service, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Lahiru Russell
- School of Nursing and Midwifery, Institute for Health Transformation, Deakin University, Geelong, VIC 3217, Australia;
- Centre for Quality and Patient Safety—Eastern Health Partnership, Box Hill, VIC 3128, Australia
| | - Victoria Dax
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
| | - Justine Diggens
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
| | - Holly Kent
- Department of Psychology and Consultation Liaison Psychiatry, Alfred Health, Melbourne, VIC 3004, Australia; (L.K.); (H.K.)
| | - Alison Button-Sloan
- Melanoma Patients Australia, Melbourne, VIC 3000, Australia;
- Melanoma Research Victoria Consumer Reference Group, Melbourne, VIC 3000, Australia
- Australian Melanoma Consumer Alliance, Melbourne, VIC 3000, Australia
| | - Jane Elliott
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
| | - Mark Shackleton
- Department of Medical Oncology, Alfred Health, Melbourne, VIC 3004, Australia; (M.S.); (H.B.)
- Department of Medicine, Monash University, Melbourne, VIC 3010, Australia
| | - Hayley Burridge
- Department of Medical Oncology, Alfred Health, Melbourne, VIC 3004, Australia; (M.S.); (H.B.)
| | - Maria Ftanou
- Psychosocial Oncology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (S.E.); (V.D.); (J.D.); (J.E.); (M.F.)
- Centre for Mental Health, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
2373
|
Board R, Smittenaar R, Lawton S, Liu H, Juwa B, Chao D, Corrie P. Metastatic melanoma patient outcomes since introduction of immune checkpoint inhibitors in England between 2014 and 2018. Int J Cancer 2020; 148:868-875. [PMID: 32838478 PMCID: PMC7821238 DOI: 10.1002/ijc.33266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
Abstract
Immune checkpoint inhibitors (CPIs) have radically changed outcomes for patients diagnosed with metastatic melanoma globally in the last 10 years, based on evidence of overall survival (OS) benefits generated from international randomised controlled trials (RCTs). Since RCTs do not always reflect real‐world prescribing, we interrogated established national databases to track prescribing of CPIs approved for first line treatment of metastatic melanoma patients in England since 2014 and determined patient outcomes associated with OS, as well as treatment‐related toxicity. Between April 2014 and March 2018, 5465 melanoma patients were diagnosed and treated with systemic anticancer therapy (SACT), 2322 of which received first‐line CPIs. There was good 3‐year OS concordance with RCT outcomes for ipilimumab (32%), ipinivo (56%) and nivolumab (51%), but OS was lower than expected for pembrolizumab (40%). Comparing patients prescribed ipinivo with those prescribed pembrolizumab, ipinivo‐treated patients were younger (88% vs 49% patients <70 years, P < .001) and fitter (60% vs 38% patients with Eastern Cooperative Oncology Group [ECOG] performance status 0, P < .0001). Emergency hospital admission rates from the earliest and last treatment dates were higher for patients prescribed ipinivo (37% and 55%) compared to those prescribed pembrolizumab (17% and 29%). The 30‐day mortality rates favoured ipinivo patients (3.8% ipinivo, 9.1% pembrolizumab, P < .0001) and likely reflected marked differences in median treatment durations: 63 (range 7‐440) days for ipinivo and 192 (range 5‐943) days for pembrolizumab. The dominant treatment‐related condition linked to hospital admission was colitis, recorded for 25% of patients prescribed ipinivo compared to 4% of patients prescribed pembrolizumab. Our population data has demonstrated that RCT outcomes can be achieved in routine care settings with careful patient selection. What's new? Immune checkpoint inhibitors (CPIs) greatly impact overall survival in metastatic melanoma. In England, CPIs have been used as first‐line therapy for this malignancy since 2014. This retrospective study assessed survival and toxicity among English metastatic melanoma patients prescribed CPIs between 2014 and 2018. For the CPIs ipilimumab, nivolumab, and ipinivo, survival outcomes were remarkably similar to registration trials. Poorer outcomes were associated with pembrolizumab, possibly because patients who received this drug were older and relatively less fit. Ipinivo generated the highest rates of emergency hospital visits and admissions, although 30‐day mortality was unchanged, potentially reflecting effective management of complications.
Collapse
Affiliation(s)
- Ruth Board
- Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Preston, UK
| | - Rebecca Smittenaar
- National Cancer Registration and Analysis Service, Health Improvement Directorate, Public Health England, London, UK
| | - Sarah Lawton
- National Cancer Registration and Analysis Service, Health Improvement Directorate, Public Health England, London, UK
| | - Hanhua Liu
- National Cancer Registration and Analysis Service, Health Improvement Directorate, Public Health England, London, UK
| | - Bukky Juwa
- National Cancer Registration and Analysis Service, Health Improvement Directorate, Public Health England, London, UK
| | - David Chao
- Department of Oncology, Royal Free London NHS Foundation Trust, London, UK
| | - Pippa Corrie
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
2374
|
Horvath L, Thienpont B, Zhao L, Wolf D, Pircher A. Overcoming immunotherapy resistance in non-small cell lung cancer (NSCLC) - novel approaches and future outlook. Mol Cancer 2020; 19:141. [PMID: 32917214 PMCID: PMC7488475 DOI: 10.1186/s12943-020-01260-z] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy (IO) has revolutionized the therapy landscape of non-small cell lung cancer (NSCLC), significantly prolonging the overall survival (OS) of advanced stage patients. Over the recent years IO therapy has been broadly integrated into the first-line setting of non-oncogene driven NSCLC, either in combination with chemotherapy, or in selected patients with PD-L1high expression as monotherapy. Still, a significant proportion of patients suffer from disease progression. A better understanding of resistance mechanisms depicts a central goal to avoid or overcome IO resistance and to improve patient outcome.We here review major cellular and molecular pathways within the tumor microenvironment (TME) that may impact the evolution of IO resistance. We summarize upcoming treatment options after IO resistance including novel IO targets (e.g. RIG-I, STING) as well as interesting combinational approaches such as IO combined with anti-angiogenic agents or metabolic targets (e.g. IDO-1, adenosine signaling, arginase). By discussing the fundamental mode of action of IO within the TME, we aim to understand and manage IO resistance and to seed new ideas for effective therapeutic IO concepts.
Collapse
MESH Headings
- Arginase/genetics
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- DEAD Box Protein 58/antagonists & inhibitors
- DEAD Box Protein 58/genetics
- DEAD Box Protein 58/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Humans
- Immunotherapy/adverse effects
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Lena Horvath
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Liyun Zhao
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
- Medical Clinic III, Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn (UKB), Sigmund-Freud-Street 25, 53127, Bonn, Germany
| | - Andreas Pircher
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
2375
|
Liu JN, Kong XS, Huang T, Wang R, Li W, Chen QF. Clinical Implications of Aberrant PD-1 and CTLA4 Expression for Cancer Immunity and Prognosis: A Pan-Cancer Study. Front Immunol 2020; 11:2048. [PMID: 33072070 PMCID: PMC7539667 DOI: 10.3389/fimmu.2020.02048] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Combination therapy with inhibitors of cytotoxic T lymphocyte-associated protein (CTLA)4 and programmed death (PD)-1 has demonstrated efficacy in cancer patients. However, there is little information on CTLA4 and PD-1 expression levels and their clinical significance across diverse cancers. In this study, we addressed this question by analyzing PD-1 and CTLA4 levels in 33 different types of cancer along with their prognostic significance using The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia datasets. Liver hepatocellular carcinoma (LIHC) patients receiving cytokine-induced killer cell (CIK) immunotherapy at Sun Yat-sen University cancer center were enrolled for survival analysis. The correlation between PD-1/CTLA4 expression and cancer immunity was also analyzed. The results showed that PD-1 and CTLA4 transcript levels varied across cancer cell lines, with aberrant expression detected in certain cancer types; Kaplan–Meier analysis with the Cox proportional hazards model showed that this was closely related to overall survival in breast invasive carcinoma, glioblastoma multiforme, head and neck squamous cell carcinoma, acute myeloid leukemialymphoma, uterine corpus endometrial carcinoma, and uveal melanoma in TCGA. High serum PD-1 and CTLA4 levels predicted better survival in LIHC patients receiving CIK therapy. PD-1 and CTLA4 levels were found to be significantly correlated with the degree of tumor cell infiltration using Tumor Immune Estimation Resource, Estimating Relative Subsets of RNA Transcripts, and Estimation of Stromal and immune Cells in Malignant Tumor Tissues Using Expression Data as well as with tumor-infiltrating lymphocyte marker expression; they were also related to tumor mutation burden, microsatellite instability, mismatch repair, and the expression of DNA methyltransferases in some cancer types. Gene set enrichment analysis of 33 cancer types provided further evidence for associations between PD-1/CTLA4 levels and cancer development and immunocyte infiltration. Thus, PD-1 and CTLA4 play important roles in tumorigenesis and tumor immunity and can serve as prognostic biomarkers in different cancer types.
Collapse
Affiliation(s)
- Jian-Nan Liu
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiang-Shuo Kong
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Tao Huang
- Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Rui Wang
- Department of Respiratory Oncology, Fushan District People's Hospital, Yantai, China
| | - Wang Li
- Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi-Feng Chen
- Department of Medical Imaging and Interventional Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
2376
|
Massari F, Mollica V, Rizzo A, Cosmai L, Rizzo M, Porta C. Safety evaluation of immune-based combinations in patients with advanced renal cell carcinoma: a systematic review and meta-analysis. Expert Opin Drug Saf 2020; 19:1329-1338. [PMID: 32799582 DOI: 10.1080/14740338.2020.1811226] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Immune-based combinations, including nivolumab plus ipilimumab, pembrolizumab plus axitinib, and (at a lesser extent) avelumab plus axitinib, should be regarded among the new standards of care for first line therapy of metastatic renal cell carcinoma. Toxicity profiles are different among all these above combinations, as well as between them and targeted agents monotherapies, including sunitinib (i.e. the control arm of all the above studies). AREAS COVERED We performed a systematic review and meta-analysis with the aim to compare adverse events from immune-based combinations versus sunitinib monotherapy across four recent randomized controlled trials (CheckMate-214, Keynote-426, IMmotion-151, and JAVELIN Renal 101) of front-line treatment for metastatic renal cell carcinoma, with particular attention to those from the ipilimumab plus nivolumab combination. EXPERT OPINION Beyond efficacy and activity, the ipilimumab plus nivolumab combination appears feasible, being endowed by an acceptable safety profile, in line with that of the other available options for the treatment of metastatic RCC. The different patterns of toxicities emerging from this systematic review and meta-analysis need to be kept in mind while choosing the appropriate treatment for each individual patient. Furthermore, prevention, prompt identification, and treatment of immune-related adverse events remains an area to be improved.
Collapse
Affiliation(s)
- Francesco Massari
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Veronica Mollica
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Rizzo
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Laura Cosmai
- Onco-Nephrology Outpatients Clinic, Division of Nephrology and Dialysis, ASST Santi Paolo e Carlo, San Paolo Hospital , Milan, Italy
| | - Mimma Rizzo
- Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri , Pavia, Italy
| | - Camillo Porta
- Chair of Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari 'A. Moro' , Bari, Italy
| |
Collapse
|
2377
|
Hu H, She L, Liao M, Shi Y, Yao L, Ding D, Zhu Y, Zeng S, Carbone DP, Huang J. Cost-Effectiveness Analysis of Nivolumab Plus Ipilimumab vs. Chemotherapy as First-Line Therapy in Advanced Non-Small Cell Lung Cancer. Front Oncol 2020; 10:1649. [PMID: 33014826 PMCID: PMC7507990 DOI: 10.3389/fonc.2020.01649] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023] Open
Abstract
Background: The CheckMate 227 trial has indicated that nivolumab plus ipilimumab compared with chemotherapy significantly increases long-term survival in the first-line setting of advanced non-small-cell lung cancer (NSCLC). Methods: A Markov model was built to estimate the cost and effectiveness of nivolumab plus ipilimumab vs. chemotherapy as the first-line therapy in patients with advanced NSCLC based on outcomes data from the CheckMate 227 trial. We calculated the cost and health outcomes at a willingness-to-pay (WTP) threshold of $150,000 per quality adjusted life year (QALY) in populations with different programmed death ligand 1 (PD-L1) expression levels (≥50, ≥1, and <1%) or a high tumor mutational burden (TMB) (≥10 mutations per megabase). Sensitivity analysis were used to test the model stability. Results: The outcomes showed that the incremental costs and QALYs by using nivolumab plus ipilimumab were $124180.76 and 1.16, $70951.42 and 0.53, $144093.63 and 0.83 for the advanced NSCLC patients with a PD-L1 expression ≥50%, ≥1%, and <1%, which led to an incremental cost-effective ratio (ICER) of $107403.72, $133732.20, and $172589.15 per QALY, respectively. For patients with a high TMB, nivolumab plus ipilimumab contributed an extra 2.04 QALYs at a cost of $69182.50 per QALY. Conclusion: Nivolumab plus ipilimumab as first-line therapy makes a better cost-effective strategy than chemotherapy in advanced NSCLC patients with PD-L1 expression levels ≥50% and ≥1% or a high TMB, at a willingness-to-pay threshold of $150,000 per QALY, but not in the patients with a PD-L1 expression <1%.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Longjiang She
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Mengting Liao
- Xiangya Hospital, Central South University, Changsha, China
| | - Yin Shi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Linli Yao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Dong Ding
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youwen Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - David P Carbone
- Barbara J. Bonner Chair in Lung Cancer Research, James Thoracic Center, James Cancer Center, The Ohio State University Medical Center, Columbus, OH, United States
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2378
|
Ascierto PA, Puzanov I, Agarwala SS, Blank C, Carvajal RD, Demaria S, Dummer R, Ernstoff M, Ferrone S, Fox BA, Gajewski TF, Garbe C, Hwu P, Lo RS, Long GV, Luke JJ, Osman I, Postow MA, Sullivan RJ, Taube JM, Trinchieri G, Zarour HM, Caracò C, Thurin M. Perspectives in melanoma: meeting report from the "Melanoma Bridge" (December 5th-7th, 2019, Naples, Italy). J Transl Med 2020; 18:346. [PMID: 32894202 PMCID: PMC7487701 DOI: 10.1186/s12967-020-02482-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
The melanoma treatment landscape changed in 2011 with the approval of the first anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4 checkpoint inhibitor and of the first BRAF-targeted monoclonal antibody, both of which significantly improved overall survival (OS). Since then, improved understanding of the tumor microenvironment (TME) and tumor immune-evasion mechanisms has resulted in new approaches to targeting and harnessing the host immune response. The approval of new immune and targeted therapies has further improved outcomes for patients with advanced melanoma and other combination modalities are also being explored such as chemotherapy, radiotherapy, electrochemotherapy and surgery. In addition, different strategies of drugs administration including sequential or combination treatment are being tested. Approaches to overcome resistance and to potentiate the immune response are being developed. Increasing evidence emerges that tissue and blood-based biomarkers can predict the response to a therapy. The latest findings in melanoma research, including insights into the tumor microenvironment and new biomarkers, improved understanding of tumor immune response and resistance, novel approaches for combination strategies and the role of neoadjuvant and adjuvant therapy, were the focus of discussions at the Melanoma Bridge meeting (5-7 December, 2019, Naples, Italy), which are summarized in this report.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Via Mariano Semmola, 80131, Naples, Italy.
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | - Richard D Carvajal
- Columbia University Irving Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Reinhard Dummer
- Department of Dermatology, University of Zurich Hospital, Zurich, Switzerland
| | - Marc Ernstoff
- Roswell Park Comprehensive Cancer Center, Jacobs School of Medicine and Biomedical Sciences, State University, Buffalo, NY, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Cancer Institute, Portland, OR, USA
| | - Thomas F Gajewski
- Department of Pathology, University of Chicago, Chicago, IL, USA
- Department of Medicine (Section of Haematology/Oncology), University of Chicago, Chicago, IL, USA
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, Anderson Cancer Center, Houston, TX, USA
| | - Roger S Lo
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney and Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Jason J Luke
- Medicine University of Chicago, Chicago, IL, USA
| | - Iman Osman
- The Interdisciplinary Melanoma Program, New York University Langone Medical Center, NYU Grossman Medical School, New York, NY, USA
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Ryan J Sullivan
- Melanoma Program, Mass General Cancer Center, Boston, MA, USA
| | - Janis M Taube
- Division of Dermatopathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hassane M Zarour
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Corrado Caracò
- Department Melanoma, Soft Tissue, Muscle-Skeletal and Head-Neck, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Magdalena Thurin
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, MD, USA
| |
Collapse
|
2379
|
Hultin S, Nahar K, Menzies AM, Long GV, Fernando SL, Atkinson V, Cebon J, Wong MG. Histological diagnosis of immune checkpoint inhibitor induced acute renal injury in patients with metastatic melanoma: a retrospective case series report. BMC Nephrol 2020; 21:391. [PMID: 32894101 PMCID: PMC7487459 DOI: 10.1186/s12882-020-02044-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have become the standard of care in many oncological conditions but are associated with a spectrum of renal immune-related adverse events (IrAEs). We aimed to describe the spectrum, histology, management and outcomes of renal IrAE in patients with metastatic melanoma undergoing ICI therapy. METHODS We conducted a retrospective review of 23 patients with a diagnosis of metastatic melanoma treated with ICI between January 2017 and April 2019 who developed a renal IrAE. Baseline demographic data, biochemical and histopathological results, management and outcomes were analyzed. RESULTS The majority of patients who developed renal irAE were male and received combination immunotherapy. The median time of onset from initiation of ICI therapy to renal IrAE was 4 months. 52% of the treated renal IrAE had histopathologically confirmed renal IrAE. The most common histological pattern of injury was acute tubulo-interstitial nephritis (92%). One patient developed anti-GBM disease with non-dialysis dependent stage 5 CKD. In tubulointerstitial injury, there was no association between peak creatinine, renal recovery and histologically reported inflammation or fibrosis. Patients with renal IrAE demonstrated persisting renal dysfunction at 3, 6 and 12 months with a mean baseline, 3 and 12 month creatinine of 90.0 μmol/L, 127.0 μmol/L and 107.5 μmol/L respectively. CONCLUSION Renal IrAE is most commonly attributable to steroid responsive acute tubulointerstitial nephritis. The outcome of rarer pathologies such as anti-GBM disease may be adversely affected by a delayed diagnosis. There is persisting renal dysfunction following an episode of renal IrAE that may have impact on future renal and overall survival outcomes.
Collapse
Affiliation(s)
- Sebastian Hultin
- Department of Renal Medicine Royal North Shore Hospital, Sydney, Australia. .,Sydney Medical School, The University of Sydney, Sydney, Australia. .,Westmead Institute of Medical Research, 176 Hawkesbury Road, Westmead, NSW2145, Australia.
| | - Kazi Nahar
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Royal North Shore Hospital and Mater Hospital, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Royal North Shore Hospital and Mater Hospital, Sydney, Australia
| | - Suran L Fernando
- Sydney Medical School, The University of Sydney, Sydney, Australia.,Department of Immunopathology, NSW Health Pathology North, Sydney, Australia
| | - Victoria Atkinson
- Princess Alexandra Hospital, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute at Austin Health, Heidelberg, Australia
| | - Muh Geot Wong
- Department of Renal Medicine Royal North Shore Hospital, Sydney, Australia.,The George Institute for Global Health, Sydney, Australia
| |
Collapse
|
2380
|
Bai X, Flaherty KT. Targeted and immunotherapies in BRAF mutant melanoma: where we stand and what to expect. Br J Dermatol 2020; 185:253-262. [PMID: 32652567 DOI: 10.1111/bjd.19394] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2020] [Indexed: 01/01/2023]
Abstract
The therapeutic landscape for melanoma has evolved drastically in the past decade. Currently, immune checkpoint inhibitors and small-molecule inhibitors targeting the mitogen-activated protein kinase (MAPK) pathway are the two mainstay therapies for BRAFV600 mutant advanced melanoma. Although MAPK dependence has been variably demonstrated in melanomas lacking BRAFV600 mutations, definitive evidence of benefit with MAPK inhibitors has not been demonstrated. Thus, in the BRAFV600 'wild-type' setting, immune checkpoint inhibitors are the standalone option(s). In the BRAFV600 mutant setting, there is no definitive evidence prioritizing one therapeutic modality over another. Herein, we review the updated data of the pivotal phase III randomized controlled trials that established the standard-of-care first-line treatment for advanced melanoma, as it provides insights into long-term benefit, which is a major factor in therapy selection. We discuss the clinical considerations for choosing between these therapies in the front-line setting and beyond, specifically for patients with BRAFV600 mutant melanoma based on currently available evidence. We have previously proposed a time-dependent resistance paradigm in which future therapeutic development strategies can be rooted. We also discuss how these Food and Drug Administration (FDA)-approved therapeutic modalities are being pursued earlier in the course of disease management, namely in adjuvant and neoadjuvant settings. FDA-approved interlesional oncolytic virotherapy in the modern era is also briefly discussed.
Collapse
Affiliation(s)
- X Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China.,Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - K T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2381
|
Li M, Sack JS, Rahma OE, Hodi FS, Zucker SD, Grover S. Outcomes after resumption of immune checkpoint inhibitor therapy after high-grade immune-mediated hepatitis. Cancer 2020; 126:5088-5097. [PMID: 32888341 DOI: 10.1002/cncr.33165] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND In the current study, the authors assessed the risks and outcomes of immune checkpoint inhibitor (ICI) rechallenge in patients with resolved grade 3 to 4 ICI hepatitis because current guidelines recommend permanent ICI discontinuation in these patients. METHODS The authors performed a retrospective cohort study from 2010 through 2019 of patients with melanoma who were treated with ≥1 ICIs and who recovered from grade 3 to 4 ICI hepatitis. The primary outcome was hepatitis recurrence and the secondary outcome was the development of any immune-related adverse event (irAE) requiring the discontinuation of ICI rechallenge. Best overall response and time to all-cause death were compared between the patients who did and those who did not undergo ICI rechallenge. Grading was performed using the National Cancer Institute Common Terminology Criteria for Adverse Events (version 5.0). RESULTS Of the 102 patients with melanoma who developed high-grade ICI hepatitis, 31 underwent ICI rechallenge. Although 15 of 31 patients (48%) developed an irAE of any grade, only 6 patients (19%) required ICI discontinuation due to irAE severity (4 of 29 patients [14%] rechallenged with anti-PD-1 or anti-PD-L1 and 2 of 2 patients [100%] rechallenged with ipilimumab). Recurrent hepatitis accounted for 4 of these 6 cases. Rechallenged patients who did not require ICI discontinuation were found to be significantly less likely to receive ipilimumab rather than anti-PD-1 or anti-PD-L1 monotherapy (0% vs 33%; relative risk (RR), 0.1 [95% CI, 0.1-0.3; P = .032]) and significantly less likely to be rechallenged with their original ICI (8% vs 50%; RR, 0.2 [95% CI, 0.1-0.7; P = .038]). There was no difference noted with regard to best overall response or time to death between rechallenged and non-rechallenged patients. CONCLUSIONS ICI therapy can be resumed in patients with melanoma who have recovered from grade 3 to 4 ICI hepatitis with a modest risk of serious irAEs. It remains unclear whether ICI retreatment improves clinical outcomes.
Collapse
Affiliation(s)
- Michael Li
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jordan S Sack
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Osama E Rahma
- Harvard Medical School, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Harvard Medical School, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Stephen D Zucker
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Shilpa Grover
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2382
|
Cowey CL, Boyd M, Aguilar KM, Beeks A, Krepler C, Scherrer E. An observational study of drug utilization and associated outcomes among adult patients diagnosed with BRAF-mutant advanced melanoma treated with first-line anti-PD-1 monotherapies or BRAF/MEK inhibitors in a community-based oncology setting. Cancer Med 2020; 9:7863-7878. [PMID: 32871054 PMCID: PMC7643646 DOI: 10.1002/cam4.3312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Anti‐PD‐1 monotherapies (aPD‐1) and BRAF/MEK inhibitors (BRAF/MEKi) changed the BRAF‐mutant advanced melanoma treatment landscape. This study aimed to improve the understanding of real‐world treatment patterns and optimal treatment sequence. Methods This was a retrospective study of BRAF‐mutant advanced melanoma patients who initiated 1L aPD‐1 or BRAF/MEKi in the US Oncology Network between 1 January 2014 and 31 December 2017, followed through 31 December 2018. Patient and treatment characteristics were assessed descriptively, with Kaplan‐Meier methods used for time‐to‐event endpoints. As the primary analysis, overall survival (OS) and physician‐assessed progression‐free survival (rwPFS) were evaluated with Cox proportional hazard regression models and propensity score matching (n = 49). Results A total of 224 patients were included (median age 61 years, 62.9% male, 89.7% white): 36.2% received aPD‐1 and 63.8% BRAF/MEKi. Median OS and rwPFS were longer among aPD‐1 vs BRAF/MEKi patients (OS: not reached vs 13.9 months, log‐rank P = .0169; rwPFS: 7.6 vs 6.5 months, log‐rank P = .0144). Receipt of aPD‐1 was associated with improved OS (HR = 0.602 vs BRAF/MEKi [95%CI 0.382‐0.949]; P = .0287). Among patients without an event within 6 months of 1L initiation, receipt of aPD‐1 was associated with a decreased risk of progression or death from 6 months onwards (HR = 0.228 [95%CI 0.106‐0.493]; P = .0002). This association was not observed among patients within 6 months of 1L initiation (HR = 1.146; 95% CI 0.755‐1.738). Results from the propensity score‐matched pairs were consistent with these trends. Conclusion These results suggest a clinical benefit of 1L aPD‐1 compared to BRAF/MEKi after 6 months of treatment for BRAF‐mutant advanced melanoma. Future research should explore factors associated with early progression and their relationship with clinical outcomes.
Collapse
Affiliation(s)
- Charles L Cowey
- Texas Oncology Baylor Charles A Sammons Cancer Center, Dallas, TX, USA.,McKesson Life Sciences, The Woodlands, TX, USA
| | - Marley Boyd
- McKesson Life Sciences, The Woodlands, TX, USA
| | | | - April Beeks
- McKesson Life Sciences, The Woodlands, TX, USA
| | | | | |
Collapse
|
2383
|
Watson GA, Doi J, Hansen AR, Spreafico A. Novel strategies in immune checkpoint inhibitor drug development: How far are we from the paradigm shift? Br J Clin Pharmacol 2020; 86:1753-1768. [PMID: 32394468 PMCID: PMC7444803 DOI: 10.1111/bcp.14355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/11/2022] Open
Abstract
The development of immune checkpoint inhibitors (ICI) represents a major milestone in immune-oncology. Over the years these agents have demonstrated efficacy in an increasing array of malignancies. Despite this success however, significant challenges remain. Novel approaches to both drug development and trial design are required to incorporate the unique pharmacokinetic and pharmacodynamic properties of ICIs. Further, it has also been established that the benefit of ICIs is limited to only a subset of patients. The molecular interactions between native immune cells and tumorigenesis and progression represent an active area of biomarker research, and elucidating the mechanisms of response and resistance is crucial to develop rational trial designs for the next wave of immune-oncology (IO) clinical trials, particularly in patients with primary and/or acquired resistance. Efforts are now being made to integrate both biological and clinical information using novel multi-omic approaches which are now being developed to further elucidate the molecular signatures associated with IO treatment response and resistance and enable rational drug development and trial design processes. As such, precision IO and the ability to deliver patient-specific choices for ICI monotherapies or combination therapies has become an increasingly tangible goal. We herein describe the current landscape in ICI drug development and discuss the challenges and future directions in this exciting and evolving era in immune-oncology.
Collapse
Affiliation(s)
- Geoffrey Alan Watson
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Jeffrey Doi
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Aaron Richard Hansen
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| | - Anna Spreafico
- Bras Drug Development Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer CenterUniversity Health NetworkTorontoONCanada
| |
Collapse
|
2384
|
Bisschop C, de Heer EC, Brouwers AH, Hospers GAP, Jalving M. Rational use of 18F-FDG PET/CT in patients with advanced cutaneous melanoma: A systematic review. Crit Rev Oncol Hematol 2020; 153:103044. [PMID: 32673997 DOI: 10.1016/j.critrevonc.2020.103044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/13/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is increasingly used in patients with advanced melanoma. Immune checkpoint inhibitors and BRAF/MEK-targeted therapy have transformed the therapeutic landscape of metastatic melanoma. Consequently, a need for markers predicting (early) response to treatment and for monitoring treatment (toxicity) has arisen. This systematic review appraises the current literature evidence for rational use of 18F-FDG PET/CT scans in staging, clinical decision-making, treatment monitoring and follow-up in advanced melanoma. 18F-FDG PET/CT has high overall accuracy for detection of distant metastases and is, combined with cerebral MRI, the preferred imaging strategy for staging metastatic melanoma. In contrast, strong evidence supporting the standard use of 18F-FDG PET/CT for predicting and monitoring therapy response and toxicity is currently lacking. Essential for determining the position of 18F-FDG PET/CT during treatment course in advanced melanoma are well-designed studies with standardized scanning protocols, incorporation of clinical parameters and comparison with contrast-enhanced CT alone.
Collapse
Affiliation(s)
- C Bisschop
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - E C de Heer
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - A H Brouwers
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands
| | - G A P Hospers
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - M Jalving
- University of Groningen, University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands.
| |
Collapse
|
2385
|
Letourneur D, Danlos FX, Marabelle A. Chemokine biology on immune checkpoint–targeted therapies. Eur J Cancer 2020; 137:260-271. [DOI: 10.1016/j.ejca.2020.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022]
|
2386
|
Jia Y, Liu L, Shan B. Future of immune checkpoint inhibitors: focus on tumor immune microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1095. [PMID: 33145314 PMCID: PMC7575936 DOI: 10.21037/atm-20-3735] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy has become a powerful clinical strategy in cancer treatment. Immune checkpoint inhibitors (ICIs) have opened a new era for cancer immunotherapy. Nowadays, the number of immunotherapy drug approvals has increased, with numerous treatment options in clinical and preclinical development. However, there remain some obstacles to improve the efficacy of ICIs further. The tumor immune microenvironment (TIME) consists of cancer cell, immune cells and cytokines, et cetera. The dynamics of TIME determine the efficacies of ICIs. Although the ICIs showed manageable toxicity, immune-related adverse effects (irAEs) are still unignorable for clinicians. Since some primary resistance mechanisms exist in TIME, ICIs can only show effects in individual cancer patients. Even for the patients who responded, acquired resistance will occur to neutralize the effect of ICIs. Understanding how to increase the response rates and overcome the resistance to various classes of ICIs is the key to improving clinical efficacy. Besides the novel ICIs in development, there are some approaches to establish combination therapies are underway to improve further the efficacies of ICIs in treating cancer patients. Here, we describe the complicated TIME and state quo of ICIs to prospect the future of ICIs in cancer treatment.
Collapse
Affiliation(s)
- Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2387
|
Leonardi GC, Candido S, Falzone L, Spandidos DA, Libra M. Cutaneous melanoma and the immunotherapy revolution (Review). Int J Oncol 2020; 57:609-618. [PMID: 32582963 PMCID: PMC7384846 DOI: 10.3892/ijo.2020.5088] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
In a relatively short period of time, treatment strategies for metastatic melanoma have radically changed leading to an unprecedented improvement in patient survival. In this period, immunotherapy options have evolved from cytokine‑based approaches to antibody‑mediated inhibition of immune checkpoints, cancer vaccines and pharmacological modulation of the melanoma microenvironment. Combination of immunotherapy strategies and the association of immune checkpoint inhibitors (ICIs) with BRAF V600 targeted therapy show encouraging results. The future of drug development in this field is promising. The comprehension of primary and acquired resistance mechanisms to ICIs and the dissection of melanoma immunobiology will be instrumental for the development of new treatment strategies and to improve clinical trial design. Moreover, biomarker discovery will help patient stratification and management during immunotherapy treatment. In this review, we summarize landmark clinical trials of immune checkpoint inhibitors in advanced melanoma and discuss the rational for immunotherapy combinations. Immunotherapy approaches at early stage of clinical development and recent advances in melanoma immunotherapy biomarker development are also discussed.
Collapse
Affiliation(s)
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, I-95123 Catania
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori 'Fondazione G. Pascale', I-80131 Naples, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 70013 Heraklion, Greece
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, I-95123 Catania
| |
Collapse
|
2388
|
van der Weyden L, Brenn T, Patton EE, Wood GA, Adams DJ. Spontaneously occurring melanoma in animals and their relevance to human melanoma. J Pathol 2020; 252:4-21. [PMID: 32652526 PMCID: PMC7497193 DOI: 10.1002/path.5505] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
In contrast to other cancer types, melanoma incidence has been increasing over the last 50 years, and while it still represents less than 5% of all cutaneous malignancies, melanoma accounts for the majority of skin cancer deaths, due to its propensity to metastasise. Whilst melanoma most commonly affects the skin, it can also arise in mucosal surfaces, the eye, and the brain. For new therapies to be developed, a better understanding of the genetic landscape, signalling pathways, and tumour-microenvironmental interactions is needed. This is where animal models are of critical importance. The mouse is the foremost used model of human melanoma. Arguably this is due to its plethora of benefits as a laboratory animal; however, it is important to note that unlike humans, melanocytes are not present at the dermal-epidermal junction in mice and mice do not develop melanoma without genetic manipulation. In contrast, there are numerous reports of animals that spontaneously develop melanoma, ranging from sharks and parrots to hippos and monkeys. In addition, several domesticated and laboratory-bred animals spontaneously develop melanoma or UV-induced melanoma, specifically, fish, opossums, pigs, horses, cats, and dogs. In this review, we look at spontaneously occurring animal 'models' of melanoma and discuss their relevance to the different types of melanoma found in humans. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland..
Collapse
Affiliation(s)
| | - Thomas Brenn
- Arnie Charbonneau Cancer InstituteUniversity of CalgaryCalgaryALCanada
| | - E Elizabeth Patton
- MRC Human Genetics UnitThe MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General HospitalEdinburghUK
| | - Geoffrey A Wood
- Ontario Veterinary CollegeUniversity of GuelphGuelphONCanada
| | - David J Adams
- Wellcome Sanger InstituteWellcome Genome CampusCambridgeUK
| |
Collapse
|
2389
|
Lacal PM, Atzori MG, Ruffini F, Scimeca M, Bonanno E, Cicconi R, Mattei M, Bernardini R, D'Atri S, Tentori L, Graziani G. Targeting the vascular endothelial growth factor receptor-1 by the monoclonal antibody D16F7 to increase the activity of immune checkpoint inhibitors against cutaneous melanoma. Pharmacol Res 2020; 159:104957. [PMID: 32485280 DOI: 10.1016/j.phrs.2020.104957] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/01/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
The vascular endothelial growth factor receptor-1 (VEGFR-1) is a membrane receptor for VEGF-A, placenta growth factor (PlGF) and VEGF-B that plays a crucial role in melanoma invasiveness, vasculogenic mimicry and tumor-associated angiogenesis. Furthermore, activation of VEGFR-1 is involved in the mobilization of myeloid progenitors from the bone marrow that infiltrate the tumor. Myeloid-derived suppressor cells and tumor-associated macrophages have been involved in tumor progression and resistance to cancer treatment with immune checkpoint inhibitors (ICIs). We have recently demonstrated that the anti-VEGFR-1 monoclonal antibody (mAb) D16F7 developed in our laboratories is able to inhibit melanoma growth in preclinical in vivo models and to reduce monocyte/macrophage progenitor mobilization and tumor infiltration by myeloid cells. Aim of the study was to investigate whether the anti-VEGFR-1 mAb D16F7 affects the activity of protumoral M2 macrophages in vitro in response to PlGF and inhibits the recruitment of these cells to the melanoma site in vivo. Finally, we tested whether, through its multi-targeted action, D16F7 mAb might increase the efficacy of ICIs against melanoma. The results indicated that VEGFR-1 expression is up-regulated in human activated M2 macrophages compared to activated M1 cells and exposure to the D16F7 mAb decreases in vitro chemotaxis of activated M2 macrophages. In vivo treatment with the anti-VEGFR-1 mAb D16F7 of B6D2F1 mice injected with syngeneic B16F10 melanoma cells resulted in tumor growth inhibition associated with the modification of tumor microenvironment that involves a decrease of melanoma infiltration by M2 macrophages and PD-1+ and FoxP3+ cells. These alterations result in increased M1/M2 and CD8+/FoxP3+ ratios, which favor an antitumor and immunostimulating milieu. Accordingly, D16F7 mAb increased the antitumor activity of the ICIs anti-CTLA-4 and anti-PD-1 mAbs. Overall, these data reinforce the role of VEGFR-1-mediated-signalling as a valid target for reducing tumor infiltration by protumoral macrophages and for improving the efficacy of immunotherapy with ICIs.
Collapse
Affiliation(s)
- Pedro Miguel Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Federica Ruffini
- Laboratory of Molecular Oncology, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy; Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122, Milan, Italy; Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Rosella Cicconi
- "Centro di Servizi Interdipartimentale - Stazione per la Tecnologia Animale", University of Rome Tor Vergata, Italy
| | - Maurizio Mattei
- "Centro di Servizi Interdipartimentale - Stazione per la Tecnologia Animale", University of Rome Tor Vergata, Italy; Department of Biology, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Roberta Bernardini
- "Centro di Servizi Interdipartimentale - Stazione per la Tecnologia Animale", University of Rome Tor Vergata, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
2390
|
Moyers JT, Patel A, Shih W, Nagaraj G. Association of Sociodemographic Factors With Immunotherapy Receipt for Metastatic Melanoma in the US. JAMA Netw Open 2020; 3:e2015656. [PMID: 32876684 PMCID: PMC7489862 DOI: 10.1001/jamanetworkopen.2020.15656] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE Strides to improve survival in metastatic melanoma have been made with the use of immunotherapeutic agents in the form of immune checkpoint inhibitors. OBJECTIVE To examine the factors associated with immunotherapy receipt in patients with metastatic melanoma in the era of immune checkpoint inhibitors and the Patient Protection and Affordable Care Act. DESIGN, SETTING, AND PARTICIPANTS This cohort study used data on 9882 patients with metastatic melanoma diagnosed from January 1, 2013, to December 31, 2016, from the National Cancer Database. Patients who did not have documentation regarding immunotherapy receipt were excluded. Data analysis was performed from July 1, 2019, to December 15, 2019. EXPOSURE Receipt of immunotherapy. MAIN OUTCOMES AND MEASURES The primary outcome was the association of receipt of immunotherapy as first-line therapy with sociodemographic factors. The secondary outcome was overall survival by receipt of immunotherapy. RESULTS A total of 9512 patients (mean [SD] age, 65.1 [14.4] years; 6481 [68.1%] male; 9217 [96.9%] White) met the criteria for treatment analysis. A total of 3428 (36.0%) received immunotherapy, and 6084 (64.0%) did not. Increasing age (odds ratio [OR], 0.98; 95% CI, 0.97-0.98; P < .001) and increasing Charlson-Deyo comorbidity index (OR, 0.86; 95% CI, 0.80-0.92; P < .001) were associated with lower odds of receiving immunotherapy on regression analysis. Diagnosis in Medicaid expansion states (OR, 1.16; 95% CI, 1.05-1.27; P = .003), treatment at an academic or integrated cancer network program (OR, 1.59; 95% CI, 1.45-1.75; P < .001), and residence within the highest quartile of high school graduation rate zip code area (OR, 1.31; 95% CI, 1.09-1.56; P = .003) were associated with an increased likelihood of receiving immunotherapy. Median overall survival was 10.1 months (95% CI, 9.6-10.6 months) among all patients. Patients who received first-line immunotherapy had a median overall survival of 18.4 months (95% CI, 16.6-20.1 months) compared with 7.5 months (95% CI, 7.0-7.9 months) (P < .001) among patients who did not. CONCLUSIONS AND RELEVANCE In this cohort study, patients who received immunotherapy for metastatic melanoma had improved overall survival. Residence in Medicaid expansion states, younger age, low comorbidity index, care at academic medical centers or integrated network cancer programs, and residence in zip codes within the highest quartile of high school graduation were associated with an increased likelihood of receiving immunotherapy. Recognizing sociodemographic associations with treatment receipt is important to identify potential barriers to treatment.
Collapse
Affiliation(s)
- Justin T. Moyers
- Division of Hematology and Oncology, Department of Internal Medicine, Loma Linda University, Loma Linda, California
| | - Amie Patel
- Department of Internal Medicine, Loma Linda University, Loma Linda, California
| | - Wendy Shih
- School of Public Heath, Loma Linda University, Loma Linda, California
| | - Gayathri Nagaraj
- Division of Hematology and Oncology, Department of Internal Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
2391
|
Berardi R, Goteri G, Brunelli A, Pagliaretta S, Paolucci V, Caramanti M, Rinaldi S, Refai M, Pompili C, Morgese F, Torniai M, Marcantognini G, Ricci G, Mazzanti P, Onofri A, Bianchi F, Sabbatini A, Cascinu S. Prognostic relevance of programmed cell death protein 1/programmed death-ligand 1 pathway in thymic malignancies with combined immunohistochemical and biomolecular approach. Expert Opin Ther Targets 2020; 24:937-943. [PMID: 32662701 DOI: 10.1080/14728222.2020.1790529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 06/29/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of the study was to investigate Programmed cell Death protein 1 (PD-1) and Programmed Death-Ligand 1 (PD-L1) and their mRNA expression in thymic epithelial tumors (TETs). RESEARCH DESIGN AND METHODS We analyzed 68 samples of formalin-fixed paraffin-embedded tissue (63 thymomas and 5 thymic carcinomas). PD-1 and PD-L1 protein expression were evaluated by immunohistochemistry, and mRNA expression was evaluated by real-time PCR. RESULTS M/F ratio was 33/35, and median age was 60.5 years. Twenty patients had Myasthenia Gravis (MG). In the subgroup with large tumors (>5 cm), PD-L1 mRNA overexpression was significantly associated with worse prognosis vs. patients with no mRNA overexpression (p = 0.0083) and simultaneous PD-L1 immunostaining (>1%); PD-L1 mRNA overexpression was significantly associated with worse prognosis, respect to patient with PD-L1 negative immunostaining, and no PD-L1 mRNA overexpression (p = 0.0178). The elderly patients (>60 years) with large tumors showed worse prognosis (p = 0.0395). PD-L1 immunostaining (>50%) resulted to be significantly associated with MG. CONCLUSIONS Our data suggest the potential involvement of the PD-1 and PD-L1 pathway in TETs' progression. According to our results, it may be helpful to design future trials with anti-PD-1 drugs to establish high-risk patients after surgery.
Collapse
Affiliation(s)
- Rossana Berardi
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Gaia Goteri
- Section of Pathological Anatomy and Histopathology, Università Politecnica delle Marche , Ancona, Italy
| | | | - Silvia Pagliaretta
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Vittorio Paolucci
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Miriam Caramanti
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Silvia Rinaldi
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Majed Refai
- Thoracic Surgery, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I - GM Lancisi - G Salesi , Ancona, Italy
| | - Cecilia Pompili
- Department of Thoracic Surgery, St. James's University Hospital , Leeds, UK
| | - Francesca Morgese
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Mariangela Torniai
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Giulia Marcantognini
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Giulia Ricci
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Paola Mazzanti
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Azzurra Onofri
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Francesca Bianchi
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| | - Armando Sabbatini
- Thoracic Surgery, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I - GM Lancisi - G Salesi , Ancona, Italy
| | - Stefano Cascinu
- Medical Oncology, Università Politecnica delle Marche, Ospedali Riuniti Umberto I-GM Lancisi-G Salesi , Ancona, Italy
| |
Collapse
|
2392
|
Poels K, van Leent MMT, Reiche ME, Kusters PJH, Huveneers S, de Winther MPJ, Mulder WJM, Lutgens E, Seijkens TTP. Antibody-Mediated Inhibition of CTLA4 Aggravates Atherosclerotic Plaque Inflammation and Progression in Hyperlipidemic Mice. Cells 2020; 9:E1987. [PMID: 32872393 PMCID: PMC7565685 DOI: 10.3390/cells9091987] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
T cell-driven inflammation plays a critical role in the initiation and progression of atherosclerosis. The co-inhibitory protein Cytotoxic T-Lymphocyte Associated protein (CTLA) 4 is an important negative regulator of T cell activation. Here, we studied the effects of the antibody-mediated inhibition of CTLA4 on experimental atherosclerosis by treating 6-8-week-old Ldlr-/- mice, fed a 0.15% cholesterol diet for six weeks, biweekly with 200 μg of CTLA4 antibodies or isotype control for six weeks. 18F-fluorodeoxyglucose Positron Emission Tomography-Computed Tomography showed no effect of the CTLA4 inhibition of activity in the aorta, spleen, and bone marrow, indicating that monocyte/macrophage-driven inflammation was unaffected. Correspondingly, flow cytometry demonstrated that the antibody-mediated inhibition of CTLA4 did not affect the monocyte populations in the spleen. αCTLA4 treatment induced an activated T cell profile, characterized by a decrease in naïve CD44-CD62L+CD4+ T cells and an increase in CD44+CD62L- CD4+ and CD8+ T cells in the blood and lymphoid organs. Furthermore, αCTLA4 treatment induced endothelial activation, characterized by increased ICAM1 expression in the aortic endothelium. In the aortic arch, which mainly contained early atherosclerotic lesions at this time point, αCTLA4 treatment induced a 2.0-fold increase in the plaque area. These plaques had a more advanced morphological phenotype and an increased T cell/macrophage ratio, whereas the smooth muscle cell and collagen content decreased. In the aortic root, a site that contained more advanced plaques, αCTLA4 treatment increased the plaque T cell content. The short-term antibody-mediated inhibition of CTLA4 thus accelerated the progression of atherosclerosis by inducing a predominantly T cell-driven inflammation, and resulted in the formation of plaques with larger necrotic cores and less collagen. This indicates that existing therapies that are based on αCTLA4 antibodies may promote CVD development in patients.
Collapse
Affiliation(s)
- Kikkie Poels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
| | - Mandy M. T. van Leent
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Myrthe E. Reiche
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
| | - Pascal J. H. Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
| | - Menno P. J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
| | - Willem J. M. Mulder
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80802 Munich, Germany
| | - Tom T. P. Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, 1105AZ Amsterdam, The Netherlands; (K.P.); (M.M.T.v.L.); (M.E.R.); (P.J.H.K.); (S.H.); (M.P.J.d.W.); (W.J.M.M.); (E.L.)
- Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, 1081AV Amsterdam, The Netherlands
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, 1081AV Amsterdam, The Netherlands
| |
Collapse
|
2393
|
Bartoušková M, Melichar B. Precision medicine in medical oncology: hope, disappointment and reality. Clin Chem Lab Med 2020; 58:1427-1431. [PMID: 32284441 DOI: 10.1515/cclm-2020-0231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Indexed: 11/15/2022]
Abstract
During the past 20 years, targeted therapy based on the understanding of tumor biology has been complementing or even replacing cytotoxic agents that have dominated pharmacotherapy of cancer since the conception of medical oncology. Unfortunately, the fact that targeted therapies with potential to induce cure or at least substantially prolong survival are still not available for many common solid tumors results in skepticism or even nihilism. On the one hand, biomarker research is not keeping pace with the introduction of new agents, while on the other hand, effective drugs are still not available for many potential molecular targets associated with malignant transformation and tumor progression. However, targeted therapies have already transformed the natural history and clinical outcomes not only in patients with rare malignancies like gastrointestinal stromal tumor but also with many common tumors, e.g. breast cancer, malignant melanoma, non-small cell lung cancer or renal cell carcinoma. For further advances, a multidisciplinary effort is indispensible that should, above all, involve the collaboration of medical oncology and laboratory medicine.
Collapse
Affiliation(s)
- Marie Bartoušková
- Department of Oncology, Palacký University Medical School and Teaching Hospital, Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacký University Medical School and Teaching Hospital, Olomouc, Czech Republic
| |
Collapse
|
2394
|
Bartoušková M, Hrouzková M, Čtvrtlík F, Petrová P, Rušarová N, Kučerová K, Vernerová A, Melichar B, Študentová H. Potential utilization of neopterin measurements in the assessment of pyrexia in metastatic melanoma treated with combined targeted therapy: a case report. Pteridines 2020. [DOI: 10.1515/pteridines-2020-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
In patients with metastatic melanoma the advent of targeted therapy and immune checkpoint inhibitors has transformed the management of advanced and metastatic disease, resulting in improved outcomes. Neopterin is a biomarker of immune activation increased in cancer as well as in other conditions associated with immune activation. We present a case of a patient with advanced metastatic melanoma responding to the combination targeted therapy with dabrafenib and trametinib. The treatment was complicated by a fever that was accompanied by a marked rise in serum and urinary neopterin concentrations. Present case report illustrates not only the efficacy of combined targeted therapy, but also the utilization of neopterin measurements in the diagnosis and monitoring of pyrexia in patients with metastatic malignant melanoma.
Collapse
Affiliation(s)
- Marie Bartoušková
- Department of Oncology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Michaela Hrouzková
- Department of Oncology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Filip Čtvrtlík
- Department of Radiology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Pavla Petrová
- Department of Clinical Biochemistry, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Nikol Rušarová
- Department of Oncology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Kateřina Kučerová
- Department of Analytical Chemistry, Faculty of Pharmacy , Charles University , Hradec Králové, Czech Republic
| | - Andrea Vernerová
- Department of Analytical Chemistry, Faculty of Pharmacy , Charles University , Hradec Králové, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| | - Hana Študentová
- Department of Oncology, Faculty of Medicine and Dentistry and University Hospital , Palacký University Olomouc , Czech Republic
| |
Collapse
|
2395
|
Carrié L, Virazels M, Dufau C, Montfort A, Levade T, Ségui B, Andrieu-Abadie N. New Insights into the Role of Sphingolipid Metabolism in Melanoma. Cells 2020; 9:E1967. [PMID: 32858889 PMCID: PMC7565650 DOI: 10.3390/cells9091967] [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: 07/17/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Cutaneous melanoma is a deadly skin cancer whose aggressiveness is directly linked to its metastatic potency. Despite remarkable breakthroughs in term of treatments with the emergence of targeted therapy and immunotherapy, the prognosis for metastatic patients remains uncertain mainly because of resistances. Better understanding the mechanisms responsible for melanoma progression is therefore essential to uncover new therapeutic targets. Interestingly, the sphingolipid metabolism is dysregulated in melanoma and is associated with melanoma progression and resistance to treatment. This review summarises the impact of the sphingolipid metabolism on melanoma from the initiation to metastatic dissemination with emphasis on melanoma plasticity, immune responses and resistance to treatments.
Collapse
Affiliation(s)
- Lorry Carrié
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Mathieu Virazels
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Carine Dufau
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Anne Montfort
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Thierry Levade
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
- Laboratoire de Biochimie Métabolique, CHU, 31059 Toulouse, France
| | - Bruno Ségui
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Nathalie Andrieu-Abadie
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| |
Collapse
|
2396
|
González-Montero J, Valenzuela G, Ahumada M, Barajas O, Villanueva L. Management of cancer patients during COVID-19 pandemic at developing countries. World J Clin Cases 2020; 8:3390-3404. [PMID: 32913846 PMCID: PMC7457113 DOI: 10.12998/wjcc.v8.i16.3390] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/09/2020] [Accepted: 08/12/2020] [Indexed: 02/05/2023] Open
Abstract
Cancer patient care requires a multi-disciplinary approach and multiple medical and ethical considerations. Clinical care during a pandemic health crisis requires prioritising the use of resources for patients with a greater chance of survival, especially in developing countries. The coronavirus disease 2019 crisis has generated new challenges given that cancer patients are normally not prioritised for admission in critical care units. Nevertheless, the development of new cancer drugs and novel adjuvant/neoadjuvant protocols has dramatically improved the prognosis of cancer patients, resulting in a more complex decision-making when prioritising intensive care in pandemic times. In this context, it is essential to establish an effective and transparent communication between the oncology team, critical care, and emergency units to make the best decisions, considering the principles of justice and charity. Concurrently, cancer treatment protocols must be adapted to prioritise according to oncologic response and prognosis. Communication technologies are powerful tools to optimise cancer care during pandemics, and we must adapt quickly to this new scenario of clinical care and teaching. In this new challenging pandemic scenario, multi-disciplinary work and effective communication between clinics, technology, science, and ethics is the key to optimising clinical care of cancer patients.
Collapse
Affiliation(s)
- Jaime González-Montero
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| | - Guillermo Valenzuela
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| | - Mónica Ahumada
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Basic and Clinical Oncology Department, Hospital Clinico Universidad de Chile and Clínica Dávila, Chile
| | - Olga Barajas
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Basic and Clinical Oncology Department, Hospital Clinico Universidad de Chile and Fundación Arturo López-Pérez, Chile
| | - Luis Villanueva
- Oncology Department, Hospital Clínico Universidad de Chile and Fundación Arturo López-Perez, Chile
| |
Collapse
|
2397
|
Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, Birnbaumer L, Yang Y. Reinvigorating exhausted CD8 + cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev 2020; 41:156-201. [PMID: 32844499 DOI: 10.1002/med.21727] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has revolutionized the treatment of cancer in recent years and achieved overall success and long-term clinical benefit in patients with a wide variety of cancer types. However, there is still a large proportion of patients exhibiting limited or no responses to immunotherapeutic strategy, some of which were even observed with hyperprogressive disease. One major obstacle restricting the efficacy is that tumor-reactive CD8+ T cells, which are central for tumor control, undergo exhaustion, and lose their ability to eliminate cancer cells after infiltrating into the strongly immunosuppressive tumor microenvironment. Thus, as a potential therapeutic rationale in the development of cancer immunotherapy, targeting or reinvigorating exhausted CD8+ T cells has been attracting much interest. Hitherto, both intrinsic and extrinsic mechanisms that govern CD8+ T-cell exhaustion have been explored. Specifically, the transcriptional and epigenetic landscapes have been depicted utilizing single-cell RNA sequencing or mass cytometry (CyTOF). In addition, cellular metabolism dictating the tumor-infiltrating CD8+ T-cell fate is currently under investigation. A series of clinical trials are being carried out to further establish the current strategies targeting CD8+ T-cell exhaustion. Taken together, despite the proven benefit of immunotherapy in cancer patients, additional efforts are still needed to fully circumvent limitations of exhausted T cells in the treatment. In this review, we will focus on the current cellular and molecular understanding of metabolic changes, epigenetic remodeling, and transcriptional regulation in CD8+ T-cell exhaustion and describe hypothetical treatment approaches based on immunotherapy aiming at reinvigorating exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Md Amir Hossain
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guilai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxuan Si
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qitao Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junaid Wazir
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA.,Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
2398
|
Cohen JT, Miner TJ, Vezeridis MP. Is the neutrophil-to-lymphocyte ratio a useful prognostic indicator in melanoma patients? Melanoma Manag 2020; 7:MMT47. [PMID: 32922729 PMCID: PMC7475797 DOI: 10.2217/mmt-2020-0006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/25/2020] [Indexed: 12/25/2022] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) is gaining traction as a biomarker with utility in a variety of malignancies including melanoma. Intact lymphocyte function is necessary for tumor surveillance and destruction, and neutrophils play a role in suppressing lymphocyte proliferation and in the induction of lymphocyte apoptosis. Early research in melanoma indicates that in high-risk localized melanoma, a high NLR is correlated with worse overall and disease-free survival. Similarly, in metastatic melanoma treated with both metastasectomy and immunotherapies, an elevated NLR is predictive of shortened overall survival and progression-free survival. Future studies incorporating NLR into more traditional melanoma prognostic markers while employing more granular outcomes, are needed to realize the full potential of NLR.
Collapse
Affiliation(s)
- Joshua T Cohen
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Thomas J Miner
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Michael P Vezeridis
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
2399
|
Mihic-Probst D, Reinehr M, Dettwiler S, Kolm I, Britschgi C, Kudura K, Maggio EM, Lenggenhager D, Rushing EJ. The role of macrophages type 2 and T-regs in immune checkpoint inhibitor related adverse events. Immunobiology 2020; 225:152009. [PMID: 32962812 DOI: 10.1016/j.imbio.2020.152009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitory (ICI) therapy represents a novel approach in a variety of cancers, with impressive survival benefit. With ICIs, however, a new spectrum of immune related adverse events (irAE) including life threatening hypohysitis has emerged. This autopsy study aimed to investigate inflammatory cells, PD-1 and PD-L1 expression in cases of patients who developed hypophysitis and involvement of other organs. We analysed 6 patients, who were treated with ICIs and developed hypophysitis. Two received an additional MAP-kinase inhibitor, MEK-inhibitor and cytotoxic chemotherapy. Besides the pituitary gland, all investigated adrenal glands (5/5) were affected; three cases had other organs involved (liver (2/6), thyroid (2/6), lung (1/6), myocardium (1/6), colon (1/6). The inflammatory cells of involved organs were further specified and PD1 and PDL-1 expression was analyzed using immunohistochemistry. We observed that patients treated with ICIs alone showed T-cell predominant lymphocytic infiltrates, whereas patients receiving additional therapies demonstrated an increase in B- and T-lymphocytes. Surprisingly, the dominant inflammatory population was not T-cell, but type 2 macrophages. CD25 positive T-regs were sparse or absent. Our study suggests that T cell activation is only partially responsible for irAE. ICI therapy interaction with CTLA-4, PD-1 and PDL-1 in type 2 macrophages appears to result in disturbance of their control. Furthermore, depletion of T-regs seems to contribute significantly. Our findings with simultaneous pituitary and adrenal gland involvement underlines the systemic involvement as well as the importance of monitoring cortisol levels to avoid potentially life threatening hypocortisolism.
Collapse
Affiliation(s)
- Daniela Mihic-Probst
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.
| | - Michael Reinehr
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Susanne Dettwiler
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Isabel Kolm
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Christian Britschgi
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Ken Kudura
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Ewerton Marques Maggio
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Daniela Lenggenhager
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
2400
|
Johnson DB, Reynolds KL, Sullivan RJ, Balko JM, Patrinely JR, Cappelli LC, Naidoo J, Moslehi JJ. Immune checkpoint inhibitor toxicities: systems-based approaches to improve patient care and research. Lancet Oncol 2020; 21:e398-e404. [PMID: 32758477 DOI: 10.1016/s1470-2045(20)30107-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/24/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have now been approved in numerous and diverse cancer types and combination regimens. Effective recognition and treatment of ICI toxicities, which might occur acutely, affect any organ system, and produce many distinct clinical syndromes, have emerged as essential goals of ICI management. Thus, developing robust diagnostic and management approaches for ICI toxicity across the health-care system is an urgent and unmet clinical need. In this Personal View, we describe barriers to high-quality care that have constrained the most effective management of patients with cancer receiving ICI treatment. We review education initiatives to enhance patient and physician awareness, which is necessary given the broad spectrum of ICI toxicities often experienced by patients, and assess various systems-based approaches that maximise the chances of appropriate management. In addition, we describe research pipelines that broaden evidence-based approaches and the pathobiology of these novel events. Developing effective, systematic approaches for the recognition and treatment of ICI toxicities will continue to grow in importance as these agents proliferate in cancer care.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| | | | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Laura C Cappelli
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Javid J Moslehi
- Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|