1351
|
Klemen ND, Shindorf ML, Sherry RM. Role of Surgery in Combination with Immunotherapy. Surg Oncol Clin N Am 2019; 28:481-487. [DOI: 10.1016/j.soc.2019.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
1352
|
Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson VG, Dalle S, Haydon A, Lichinitser M, Khattak A, Carlino MS, Sandhu S, Larkin J, Puig S, Ascierto PA, Rutkowski P, Schadendorf D, Koornstra R, Hernandez-Aya L, Di Giacomo AM, van den Eertwegh AJ, Grob JJ, Gutzmer R, Jamal R, Lorigan PC, Lupinacci R, Krepler C, Ibrahim N, Kicinski M, Marreaud S, van Akkooi AC, Suciu S, Robert C. Prognostic and predictive value of AJCC-8 staging in the phase III EORTC1325/KEYNOTE-054 trial of pembrolizumab vs placebo in resected high-risk stage III melanoma. Eur J Cancer 2019; 116:148-157. [PMID: 31200321 DOI: 10.1016/j.ejca.2019.05.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND The American Joint Committee on Cancer-8 (AJCC) classification of melanoma was implemented in January 2018. It was based on data gathered when checkpoint inhibitors were not used as adjuvant therapy in stage III melanoma. The European Organization for Research and Treatment of Cancer (EORTC) 1325/KEYNOTE-054 double-blind phase III trial evaluated pembrolizumab vs placebo in AJCC-7 stage IIIA (excluding lymph node metastasis ≤1 mm), IIIB or IIIC (without in-transit metastasis) patients after complete lymphadenectomy. PATIENTS, METHODS AND RESULTS Patients (n = 1019) were randomised 1:1 to pembrolizumab 200 mg or placebo every 3 weeks (total of 18 doses, ∼1 year). At 1.25-year median follow-up, pembrolizumab prolonged relapse-free survival (RFS) in the total population (1-year RFS rate: 75.4% vs 61.0%; hazard ratio [HR] 0.57; logrank P < 0.0001) and consistently in the AJCC-7 subgroups. Prognostic and predictive values of AJCC-8 for RFS were evaluated in this study. Patient distribution according to the AJCC-8 stage subgroups was 8% (IIIA), 34.7% (IIIB), 49.7% (IIIC), 3.7% (IIID) and 3.8% (unknown). AJCC-8 classification was strongly associated with RFS (HRs for stage IIIB, IIIC and IIID vs IIIA were 4.0, 5.7 and 12.2, respectively) but showed no predictive importance for the treatment comparison regarding RFS (test for interaction: P = 0.68). The 1-year RFS rate for pembrolizumab vs placebo and the HRs (99% confidence interval) within each AJCC-8 subgroup were as follows: stage IIIA (92.7% vs 92.5%; 0.76 [0.11-5.43]), IIIB (79.0% vs 65.5%; 0.59 [0.35-0.99]), IIIC (73.6% vs 53.9%; 0.48 [0.33-0.70]) and IIID (50.0% vs 33.3%; 0.69 [0.24-2.00]). CONCLUSIONS AJCC-8 staging had a strong prognostic importance for RFS but no predictive importance: the RFS benefit of pembrolizumab was observed across AJCC-8 subgroups in resected high-risk stage III melanoma patients.
Collapse
Affiliation(s)
| | - Christian U Blank
- Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Mario Mandala
- Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Mater and Royal North Shore Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Adnan Khattak
- Fiona Stanley Hospital/University of Western Australia, Perth, WA, Australia
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, Sydney, NSW, Australia
| | | | | | - Susana Puig
- Hospital Clinic Universitari de Barcelona, Barcelona, Spain
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie Institute - Oncology Center, Warsaw, Poland
| | | | - Rutger Koornstra
- Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
| | | | | | | | | | - Ralf Gutzmer
- Skin Cancer Center, Hannover Medical School, Hannover, Germany
| | - Rahima Jamal
- Centre Hospitalier de l'Université de Montréal (CHUM), Centre de recherche du CHUM, Montreal, QC, Canada
| | - Paul C Lorigan
- Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
1353
|
Neoadjuvant systemic therapy in melanoma: recommendations of the International Neoadjuvant Melanoma Consortium. Lancet Oncol 2019; 20:e378-e389. [DOI: 10.1016/s1470-2045(19)30332-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/08/2019] [Accepted: 04/18/2019] [Indexed: 12/25/2022]
|
1354
|
Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, Shen C, Duma N, Vera Aguilera J, Chintakuntlawar A, Price KA, Molina JR, Pagliaro LC, Halfdanarson TR, Grothey A, Markovic SN, Nowakowski GS, Ansell SM, Wang ML. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol 2019; 5:1008-1019. [PMID: 31021376 PMCID: PMC6487913 DOI: 10.1001/jamaoncol.2019.0393] [Citation(s) in RCA: 582] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE Programmed cell death (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors have been increasingly used in cancer therapy. Understanding the treatment-related adverse events of these drugs is critical for clinical practice. OBJECTIVE To evaluate the incidences of treatment-related adverse events of PD-1 and PD-L1 inhibitors and the differences between different drugs and cancer types. DATA SOURCES PubMed, Web of Science, Embase, and Scopus were searched from October 1, 2017, through December 15, 2018. STUDY SELECTION Published clinical trials on single-agent PD-1 and PD-L1 inhibitors with tabulated data on treatment-related adverse events were included. DATA EXTRACTION AND SYNTHESIS Trial name, phase, cancer type, PD-1 and PD-L1 inhibitor used, dose escalation, dosing schedule, number of patients, number of all adverse events, and criteria for adverse event reporting data were extracted from each included study, and bayesian multilevel regression models were applied for data analysis. MAIN OUTCOMES AND MEASURES Incidences of treatment-related adverse events and differences between different drugs and cancer types. RESULTS This systematic review and meta-analysis included 125 clinical trials involving 20 128 patients; 12 277 (66.0%) of 18 610 patients from 106 studies developed at least 1 adverse event of any grade (severity), and 2627 (14.0%) of 18 715 patients from 110 studies developed at least 1 adverse event of grade 3 or higher severity. The most common all-grade adverse events were fatigue (18.26%; 95% CI, 16.49%-20.11%), pruritus (10.61%; 95% CI, 9.46%-11.83%), and diarrhea (9.47%; 95% CI, 8.43%-10.58%). The most common grade 3 or higher adverse events were fatigue (0.89%; 95% CI, 0.69%-1.14%), anemia (0.78%; 95% CI, 0.59%-1.02%), and aspartate aminotransferase increase (0.75%; 95% CI, 0.56%-0.99%). Hypothyroidism (6.07%; 95% CI, 5.35%-6.85%) and hyperthyroidism (2.82%; 95% CI, 2.40%-3.29%) were the most frequent all-grade endocrine immune-related adverse events. Nivolumab was associated with higher mean incidences of all-grade adverse events compared with pembrolizumab (odds ratio [OR], 1.28; 95% CI, 0.97-1.79) and grade 3 or higher adverse events (OR, 1.30; 95% CI, 0.89-2.00). PD-1 inhibitors were associated with a higher mean incidence of grade 3 or higher adverse events compared with PD-L1 inhibitors (OR, 1.58; 95% CI, 1.00-2.54). CONCLUSIONS AND RELEVANCE Different PD-1 and PD-L1 inhibitors appear to have varying treatment-related adverse events; a comprehensive summary of the incidences of treatment-related adverse events in clinical trials provides an important guide for clinicians.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
- Department of Public Health Sciences, Pennsylvania State College of Medicine, Hershey
| | - Fang Yang
- Medical School of Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xinyue Qi
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Xin Wang
- Medical School of Nanjing University, Nanjing, China
| | - Xiaoxiang Guan
- Medical School of Nanjing University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chan Shen
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Narjust Duma
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jesus Vera Aguilera
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | - Axel Grothey
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- West Cancer Center, The University of Tennessee, Memphis
| | | | | | | | - Michael L. Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
1355
|
Barquet-Munoz SA, Leitao M, Pérez Montiel MD, Santiago Concha BG. Vulvar melanoma: management of primary disease and repeated recurrences. Int J Gynecol Cancer 2019; 29:1077-1081. [PMID: 31196869 PMCID: PMC7425808 DOI: 10.1136/ijgc-2019-000610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 02/05/2023] Open
Affiliation(s)
| | - Mario Leitao
- Division of Gynecology, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York City, New York, USA
| | | | | |
Collapse
|
1356
|
Sarkar RR, Gloude NJ, Schiff D, Murphy JD. Cost-Effectiveness of Chimeric Antigen Receptor T-Cell Therapy in Pediatric Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia. J Natl Cancer Inst 2019; 111:719-726. [PMID: 30551196 PMCID: PMC6624167 DOI: 10.1093/jnci/djy193] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR-T) therapy is a promising new class of cancer therapy but has a high up-front cost. We evaluated the cost-effectiveness of CAR-T therapy among pediatric patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). METHODS We built a microsimulation model for pediatric patients with relapsed/refractory B-ALL receiving either CAR-T therapy or standard of care. Outcomes included costs, quality of life (health utility), complications, and survival. We measured cost-effectiveness with the incremental cost-effectiveness ratio (ICER), with ICERs under $100 000 per quality-adjusted life-year (QALY) considered cost effective. One-way and probabilistic sensitivity analyses were used to test model uncertainty. RESULTS Compared to standard of care, CAR-T therapy increased overall cost by $528 200 and improved effectiveness by 8.18 QALYs, resulting in an ICER of $64 600/QALY. The model was sensitive to assumptions about long-term CAR-T survival, the complete remission rate of CAR-T patients, and the health utility of long-term survivors. The base model assumed a 76.0% one-year survival with CAR-T, although if this decreased to 57.8%, then CAR-T was no longer cost effective. If the complete remission rate of CAR-T recipients decreased from 81% to 56.2%, or if the health utility of disease-free survivors decreased from 0.94 to 0.66, then CAR-T was no longer cost effective. Probabilistic sensitivity analysis found that CAR-T was cost effective in 94.8% of iterations at a willingness to pay of $100 000/QALY. CONCLUSION CAR-T therapy may represent a cost-effective option for pediatric relapsed/refractory B-ALL, although longer follow-up of CAR-T survivors is required to confirm validity of these findings.
Collapse
Affiliation(s)
- Reith R Sarkar
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA
| | - Nicholas J Gloude
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rady Children’s Hospital, San Diego, CA
| | - Deborah Schiff
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rady Children’s Hospital, San Diego, CA
| | - James D Murphy
- University of California San Diego School of Medicine, UCSD School of Medicine, La Jolla, CA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA
| |
Collapse
|
1357
|
Dinnes J, Ferrante di Ruffano L, Takwoingi Y, Cheung ST, Nathan P, Matin RN, Chuchu N, Chan SA, Durack A, Bayliss SE, Gulati A, Patel L, Davenport C, Godfrey K, Subesinghe M, Traill Z, Deeks JJ, Williams HC, Cochrane Skin Cancer Diagnostic Test Accuracy Group, Cochrane Skin Group. Ultrasound, CT, MRI, or PET-CT for staging and re-staging of adults with cutaneous melanoma. Cochrane Database Syst Rev 2019; 7:CD012806. [PMID: 31260100 PMCID: PMC6601698 DOI: 10.1002/14651858.cd012806.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Melanoma is one of the most aggressive forms of skin cancer, with the potential to metastasise to other parts of the body via the lymphatic system and the bloodstream. Melanoma accounts for a small percentage of skin cancer cases but is responsible for the majority of skin cancer deaths. Various imaging tests can be used with the aim of detecting metastatic spread of disease following a primary diagnosis of melanoma (primary staging) or on clinical suspicion of disease recurrence (re-staging). Accurate staging is crucial to ensuring that patients are directed to the most appropriate and effective treatment at different points on the clinical pathway. Establishing the comparative accuracy of ultrasound, computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET)-CT imaging for detection of nodal or distant metastases, or both, is critical to understanding if, how, and where on the pathway these tests might be used. OBJECTIVES Primary objectivesWe estimated accuracy separately according to the point in the clinical pathway at which imaging tests were used. Our objectives were:• to determine the diagnostic accuracy of ultrasound or PET-CT for detection of nodal metastases before sentinel lymph node biopsy in adults with confirmed cutaneous invasive melanoma; and• to determine the diagnostic accuracy of ultrasound, CT, MRI, or PET-CT for whole body imaging in adults with cutaneous invasive melanoma:○ for detection of any metastasis in adults with a primary diagnosis of melanoma (i.e. primary staging at presentation); and○ for detection of any metastasis in adults undergoing staging of recurrence of melanoma (i.e. re-staging prompted by findings on routine follow-up).We undertook separate analyses according to whether accuracy data were reported per patient or per lesion.Secondary objectivesWe sought to determine the diagnostic accuracy of ultrasound, CT, MRI, or PET-CT for whole body imaging (detection of any metastasis) in mixed or not clearly described populations of adults with cutaneous invasive melanoma.For study participants undergoing primary staging or re-staging (for possible recurrence), and for mixed or unclear populations, our objectives were:• to determine the diagnostic accuracy of ultrasound, CT, MRI, or PET-CT for detection of nodal metastases;• to determine the diagnostic accuracy of ultrasound, CT, MRI, or PET-CT for detection of distant metastases; and• to determine the diagnostic accuracy of ultrasound, CT, MRI, or PET-CT for detection of distant metastases according to metastatic site. SEARCH METHODS We undertook a comprehensive search of the following databases from inception up to August 2016: Cochrane Central Register of Controlled Trials; MEDLINE; Embase; CINAHL; CPCI; Zetoc; Science Citation Index; US National Institutes of Health Ongoing Trials Register; NIHR Clinical Research Network Portfolio Database; and the World Health Organization International Clinical Trials Registry Platform. We studied reference lists as well as published systematic review articles. SELECTION CRITERIA We included studies of any design that evaluated ultrasound (with or without the use of fine needle aspiration cytology (FNAC)), CT, MRI, or PET-CT for staging of cutaneous melanoma in adults, compared with a reference standard of histological confirmation or imaging with clinical follow-up of at least three months' duration. We excluded studies reporting multiple applications of the same test in more than 10% of study participants. DATA COLLECTION AND ANALYSIS Two review authors independently extracted all data using a standardised data extraction and quality assessment form (based on the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2)). We estimated accuracy using the bivariate hierarchical method to produce summary sensitivities and specificities with 95% confidence and prediction regions. We undertook analysis of studies allowing direct and indirect comparison between tests. We examined heterogeneity between studies by visually inspecting the forest plots of sensitivity and specificity and summary receiver operating characteristic (ROC) plots. Numbers of identified studies were insufficient to allow formal investigation of potential sources of heterogeneity. MAIN RESULTS We included a total of 39 publications reporting on 5204 study participants; 34 studies reporting data per patient included 4980 study participants with 1265 cases of metastatic disease, and seven studies reporting data per lesion included 417 study participants with 1846 potentially metastatic lesions, 1061 of which were confirmed metastases. The risk of bias was low or unclear for all domains apart from participant flow. Concerns regarding applicability of the evidence were high or unclear for almost all domains. Participant selection from mixed or not clearly defined populations and poorly described application and interpretation of index tests were particularly problematic.The accuracy of imaging for detection of regional nodal metastases before sentinel lymph node biopsy (SLNB) was evaluated in 18 studies. In 11 studies (2614 participants; 542 cases), the summary sensitivity of ultrasound alone was 35.4% (95% confidence interval (CI) 17.0% to 59.4%) and specificity was 93.9% (95% CI 86.1% to 97.5%). Combining pre-SLNB ultrasound with FNAC revealed summary sensitivity of 18.0% (95% CI 3.58% to 56.5%) and specificity of 99.8% (95% CI 99.1% to 99.9%) (1164 participants; 259 cases). Four studies demonstrated lower sensitivity (10.2%, 95% CI 4.31% to 22.3%) and specificity (96.5%,95% CI 87.1% to 99.1%) for PET-CT before SLNB (170 participants, 49 cases). When these data are translated to a hypothetical cohort of 1000 people eligible for SLNB, 237 of whom have nodal metastases (median prevalence), the combination of ultrasound with FNAC potentially allows 43 people with nodal metastases to be triaged directly to adjuvant therapy rather than having SLNB first, at a cost of two people with false positive results (who are incorrectly managed). Those with a false negative ultrasound will be identified on subsequent SLNB.Limited test accuracy data were available for whole body imaging via PET-CT for primary staging or re-staging for disease recurrence, and none evaluated MRI. Twenty-four studies evaluated whole body imaging. Six of these studies explored primary staging following a confirmed diagnosis of melanoma (492 participants), three evaluated re-staging of disease following some clinical indication of recurrence (589 participants), and 15 included mixed or not clearly described population groups comprising participants at a number of different points on the clinical pathway and at varying stages of disease (1265 participants). Results for whole body imaging could not be translated to a hypothetical cohort of people due to paucity of data.Most of the studies (6/9) of primary disease or re-staging of disease considered PET-CT, two in comparison to CT alone, and three studies examined the use of ultrasound. No eligible evaluations of MRI in these groups were identified. All studies used histological reference standards combined with follow-up, and two included FNAC for some participants. Observed accuracy for detection of any metastases for PET-CT was higher for re-staging of disease (summary sensitivity from two studies: 92.6%, 95% CI 85.3% to 96.4%; specificity: 89.7%, 95% CI 78.8% to 95.3%; 153 participants; 95 cases) compared to primary staging (sensitivities from individual studies ranged from 30% to 47% and specificities from 73% to 88%), and was more sensitive than CT alone in both population groups, but participant numbers were very small.No conclusions can be drawn regarding routine imaging of the brain via MRI or CT. AUTHORS' CONCLUSIONS Review authors found a disappointing lack of evidence on the accuracy of imaging in people with a diagnosis of melanoma at different points on the clinical pathway. Studies were small and often reported data according to the number of lesions rather than the number of study participants. Imaging with ultrasound combined with FNAC before SLNB may identify around one-fifth of those with nodal disease, but confidence intervals are wide and further work is needed to establish cost-effectiveness. Much of the evidence for whole body imaging for primary staging or re-staging of disease is focused on PET-CT, and comparative data with CT or MRI are lacking. Future studies should go beyond diagnostic accuracy and consider the effects of different imaging tests on disease management. The increasing availability of adjuvant therapies for people with melanoma at high risk of disease spread at presentation will have a considerable impact on imaging services, yet evidence for the relative diagnostic accuracy of available tests is limited.
Collapse
Affiliation(s)
- Jacqueline Dinnes
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | | | - Yemisi Takwoingi
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | - Seau Tak Cheung
- Dudley Hospitals Foundation Trust, Corbett HospitalDepartment of DermatologyWicarage RoadStourbridgeUKDY8 4JB
| | - Paul Nathan
- Mount Vernon HospitalMount Vernon Cancer CentreRickmansworth RoadNorthwoodUKHA6 2RN
| | - Rubeta N Matin
- Churchill HospitalDepartment of DermatologyOld RoadHeadingtonOxfordUKOX3 7LE
| | - Naomi Chuchu
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | - Sue Ann Chan
- City HospitalBirmingham Skin CentreDudley RdBirminghamUKB18 7QH
| | - Alana Durack
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation TrustDermatologyHills RoadCambridgeUKCB2 0QQ
| | - Susan E Bayliss
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | - Abha Gulati
- Barts Health NHS TrustDepartment of DermatologyWhitechapelLondonUKE11BB
| | - Lopa Patel
- Royal Stoke HospitalPlastic SurgeryStoke‐on‐TrentStaffordshireUKST4 6QG
| | - Clare Davenport
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | - Kathie Godfrey
- The University of Nottinghamc/o Cochrane Skin GroupNottinghamUK
| | - Manil Subesinghe
- King's College LondonCancer Imaging, School of Biomedical Engineering & Imaging SciencesLondonUK
| | - Zoe Traill
- Oxford University Hospitals NHS TrustChurchill Hospital Radiology DepartmentOxfordUK
| | - Jonathan J Deeks
- University of BirminghamInstitute of Applied Health ResearchBirminghamUKB15 2TT
| | - Hywel C Williams
- University of NottinghamCentre of Evidence Based DermatologyQueen's Medical CentreDerby RoadNottinghamUKNG7 2UH
| | | | | |
Collapse
|
1358
|
Arnaud L, Lebrun-Vignes B, Salem JE. Checkpoint inhibitor-associated immune arthritis. Ann Rheum Dis 2019; 78:e68. [PMID: 29724725 DOI: 10.1136/annrheumdis-2018-213470] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Laurent Arnaud
- Department of Rheumatology, Hôpitaux Universitaires de Strasbourg & INSERM UMRS-1109, Strasbourg, France
| | - Bénédicte Lebrun-Vignes
- AP-HP, Pitié-Salpêtrière Hospital, Department ofPharmacology, CIC-1421, Pharmacovigilance Unit; INSERM, UMR ICAN 1166;Sorbonne Universités, UPMC Univ Paris 06, Faculty of Medicine; Institute ofCardiometabolism and Nutrition (ICAN), F-75013, Paris, France
| | - Joe-Elie Salem
- AP-HP, Pitié-Salpêtrière Hospital, Department ofPharmacology, CIC-1421, Pharmacovigilance Unit; INSERM, UMR ICAN 1166;Sorbonne Universités, UPMC Univ Paris 06, Faculty of Medicine; Institute ofCardiometabolism and Nutrition (ICAN), F-75013, Paris, France
| |
Collapse
|
1359
|
Hall ET, Singhal S, Dickerson J, Gabster B, Wong HN, Aslakson RA, Schapira L. Patient-Reported Outcomes for Cancer Patients Receiving Checkpoint Inhibitors: Opportunities for Palliative Care-A Systematic Review. J Pain Symptom Manage 2019; 58:137-156.e1. [PMID: 30905677 DOI: 10.1016/j.jpainsymman.2019.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/19/2022]
Abstract
CONTEXT Immune checkpoint inhibitors (ICIs) are increasingly used to treat a variety of cancers, but comparatively little is known about patient-reported outcomes (PROs) and health-related quality of life (HRQoL) among patients receiving these novel therapies. OBJECTIVES We performed a systematic review to examine PROs and HRQoL among cancer patients receiving ICIs as compared to other anticancer therapies. METHODS We systematically searched PubMed, CINAHL, Embase, Web of Science, and Scopus, using search terms representing ICIs, PROs, and HRQoL on August 10, 2018. Eligible articles were required to involve cancer patients treated with ICIs and to report PROs and/or HRQoL data. RESULTS We screened 1453 references and included 15 publications representing 15 randomized controlled trials in our analysis. Studies included several cancer types (melanoma, lung cancer, genitourinary cancer, and head/neck cancer), used four different ICIs (nivolumab, pembrolizumab, atezolizumab, and ipilimumab), and compared ICIs to a wide range of therapies (chemotherapy, targeted therapies, other immunotherapy strategies, and placebo). Studies used a total of seven different PROs to measure HRQOL, most commonly the European Organisation for the Research and Treatment of Cancer core quality of life questionnaire (EORTC QLQ-C30) (n = 12, 80%). PRO data were reported in a variety of formats and at a variety of time points throughout treatment, which made direct comparison challenging. Some trials (n = 11, 73%) reported PROs on specific symptoms. In general, patients receiving ICIs had similar-to-improved HRQoL and experiences when compared to other therapies. CONCLUSION Despite the broad clinical trials experience of ICI therapies across cancer types, relatively few randomized studies reported PROs and patient HRQoL data. Available data suggest that ICIs are well tolerated in terms of HRQoL compared to other anticancer therapies although the conclusions are limited by the heterogeneity of trial designs and outcomes. Currently used instruments may fail to capture important symptomatology unique to ICIs, underscoring a need for PROs designed specifically for ICIs.
Collapse
Affiliation(s)
- Evan T Hall
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, California, USA
| | - Surbhi Singhal
- Department of Medicine, Stanford University, Stanford, California, USA
| | - James Dickerson
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Brooke Gabster
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Hong-Nei Wong
- Lane Medical Library & Knowledge Management Center, Stanford University School of Medicine, Stanford, California, USA
| | - Rebecca A Aslakson
- Department of Medicine, Stanford University, Stanford, California, USA; Department of Anesthesiology, Stanford University, Stanford, California, USA
| | - Lidia Schapira
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, California, USA; Department of Medicine, Stanford University, Stanford, California, USA; Department of Anesthesiology, Stanford University, Stanford, California, USA.
| | | |
Collapse
|
1360
|
Refae S, Gal J, Brest P, Milano G. Germinal immunogenetics as a predictive factor for immunotherapy. Crit Rev Oncol Hematol 2019; 141:146-152. [PMID: 31301542 DOI: 10.1016/j.critrevonc.2019.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 01/17/2023] Open
Abstract
Clinical response to checkpoint inhibitors-based (CPIs) therapies can vary among tumor types and between patients. This led to a significant amount of pre-clinical and clinical research into biomarker identification. Biomarkers have been found to cover both the tumor itself and the tumor microenvironment. Entering host-related parameters into the equation should provide a valuable strategy for identifying not only factors predictive of treatment efficacy but also of treatment-related toxicity. It is clear that germline variants can offer efficient and easily-assessable indicators (blood DNA) to enlarge the spectrum of predictive markers for CPI-based treatment. A major issue concerns the real functional significance of the reported single-nucleotide polymorphisms (SNPs) linked to CPI-treatment outcome. Powered calculations should lead to an optimal trade-off between sample size and allele frequency. New molecular technologies and new analytical methods should provide opportunities to bridge the knowledge gap between SNP-CPI treatment associations and the functional impact of these SNPs.
Collapse
Affiliation(s)
- Sadal Refae
- Centre Antoine Lacassagne, Medical Oncology Department, University Côte d'Azur, Nice, F-06189, France
| | - Jocelyn Gal
- Centre Antoine Lacassagne, Epidemiology and Biostatistics Department, University Côte d'Azur, Nice, F-06189, France
| | - Patrick Brest
- Centre Antoine Lacassagne, Cnrs, Inserm, Ircan, FHU-Oncoage, University Côte d'Azur, Nice, F-06189, France
| | - Gerard Milano
- Centre Antoine Lacassagne, Oncopharmacology Unit, University Côte d'Azur, Nice, F-06189, France.
| |
Collapse
|
1361
|
da Silva JL, Dos Santos ALS, Nunes NCC, de Moraes Lino da Silva F, Ferreira CGM, de Melo AC. Cancer immunotherapy: the art of targeting the tumor immune microenvironment. Cancer Chemother Pharmacol 2019; 84:227-240. [PMID: 31240384 DOI: 10.1007/s00280-019-03894-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/14/2019] [Indexed: 01/07/2023]
Abstract
For many decades, cancer treatment has been strongly directed toward the development of cytotoxic and cytostatic drugs, quite often leading to disappointing results due to the inter- and intra-tumoral heterogeneity. Lately, this intra-cellular look has given way to the understanding of the tumor microenvironment, thus enabling modification of the immunological dynamics between tumor cells and their host. An era of new drugs aiming to unlock the host immune system against tumor cells is steadily increasing. Strategies involving adoptive cell therapy, therapeutic vaccines, immune checkpoint inhibitors and so on have provided spectacular clinical responses and increased survival in previously refractory settings and "hard-to-treat" cancers. Based on a comprehensive search in the main scientific databases, annals of recent renowned oncology congresses and platforms of ongoing trials, the clinical pharmacology characteristics of the main classes of immunotherapeutic agents, as well as the new treatment strategies related to immunotherapy in solid tumors, are carefully discussed throughout this review.
Collapse
Affiliation(s)
- Jesse Lopes da Silva
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil.
- Oncomed Clinic Oncologica, Niterói, Rio de Janeiro, 24220-300, Brazil.
| | | | - Natalia Cristina Cardoso Nunes
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Oncoclinicas Institute for Research and Education, Sao Paulo, Brazil
| | | | | | - Andreia Cristina de Melo
- Clinic Oncomed, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Oncoclinicas Institute for Research and Education, Sao Paulo, Brazil
| |
Collapse
|
1362
|
Abstract
OPINION STATEMENT In recent years, the number of patients with malignant melanoma has continued to increase globally; surgery remains the first treatment option for patients with resectable melanoma. Adjuvant therapy for patients with stage III and IV melanoma following surgical resection has gradually been approved. After complete resection, these patients can probably derive significant benefit from adjuvant therapy. New treatments that improve the long-term survival of patients with unresectable advanced or metastatic melanoma are currently under evaluation in adjuvant therapy to increase relapse-free survival and overall survival. We here review several relevant clinical trials of radiotherapy, systemic immune therapies, molecular-targeted therapies, and neoadjuvant therapies in order to shed light on most suitable adjuvant therapy. The findings of this review include the following: The use of interferon-α2b will be restricted for patients with ulcerated primary melanoma in countries with no access to new drugs in adjuvant therapy. Ipilimumab should not be considered as the first-line therapy due to its lower efficacy and severe toxicity. The use of anti-programmed death-1 antibody would be a relevant adjuvant therapy for patients without BRAF mutation. If the BRAF mutation status is positive, the combination of dabrafenib and trametinib is a plausible option. The establishment of appropriate therapeutic planning and clinical endpoints in adjuvant therapy should affect the standard of care. The choice of optimal adjuvant therapy for individual patients is an important issue.
Collapse
Affiliation(s)
- Maiko Wada-Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
1363
|
Perone JA, Farrow N, Tyler DS, Beasley GM. Contemporary Approaches to In-Transit Melanoma. J Oncol Pract 2019; 14:292-300. [PMID: 29746804 DOI: 10.1200/jop.18.00063] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In-transit melanoma represents a distinct disease pattern of heterogeneous superficial tumors. Many treatments have been developed specifically for this type of disease, including regional chemotherapy and a variety of directly injectable agents. Novel strategies include the intralesional delivery of oncolytic viruses and immunocytokines. The combination of intralesional or regional chemotherapy with systemic immune checkpoint inhibitors also is a promising approach. In the current review, we examine the general management of the workup of patients with in-transit disease, the range of available therapies, and recommendations for specific therapies for an individual patient.
Collapse
Affiliation(s)
- Jennifer A Perone
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Nellie Farrow
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Douglas S Tyler
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| | - Georgia M Beasley
- University Texas Medical Branch, Galveston, TX; and Duke University, Durham, NC
| |
Collapse
|
1364
|
Ellis R, Tang D, Nasr B, Greenwood A, McConnell A, Anagnostou ME, Elias M, Verykiou S, Bajwa D, Ewen T, Reynolds NJ, Barrett P, Carling E, Watson G, Armstrong J, Allen AJ, Horswell S, Labus M, Lovat PE. Epidermal autophagy and beclin 1 regulator 1 and loricrin: a paradigm shift in the prognostication and stratification of the American Joint Committee on Cancer stage I melanomas. Br J Dermatol 2019; 182:156-165. [PMID: 31056744 PMCID: PMC6973157 DOI: 10.1111/bjd.18086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND The updated American Joint Committee on Cancer (AJCC) staging criteria for melanoma remain unable to identify high-risk stage I tumour subsets. OBJECTIVES To determine the utility of epidermal autophagy and beclin 1 regulator 1 (AMBRA1)/loricrin (AMLo) expression as a prognostic biomarker for AJCC stage I cutaneous melanoma. METHODS Peritumoral AMBRA1 expression was evaluated in a retrospective discovery cohort of 76 AJCC stage I melanomas. AMLo expression was correlated with clinical outcomes up to 12 years in two independent powered, retrospective validation and qualification cohorts comprising 379 AJCC stage I melanomas. RESULTS Decreased AMBRA1 expression in the epidermis overlying primary melanomas in a discovery cohort of 76 AJCC stage I tumours was associated with a 7-year disease-free survival (DFS) rate of 81·5% vs. 100% survival with maintained AMBRA1 (P < 0·081). Following an immunohistochemistry protocol for semi-quantitative analysis of AMLo, analysis was undertaken in validation (n = 218) and qualification cohorts (n = 161) of AJCC stage I melanomas. Combined cohort analysis revealed a DFS rate of 98·3% in the AMLo low-risk group (n = 239) vs. 85·4% in the AMLo high-risk cohort (n = 140; P < 0·001). Subcohort multivariate analysis revealed that an AMLo hazard ratio (HR) of 4·04 [95% confidence interval (CI) 1·69-9·66; P = 0·002] is a stronger predictor of DFS than Breslow depth (HR 2·97, 95% CI 0·93-9·56; P = 0·068) in stage IB patients. CONCLUSIONS Loss of AMLo expression in the epidermis overlying primary AJCC stage I melanomas identifies high-risk tumour subsets independently of Breslow depth. What's already known about this topic? There is an unmet clinical need for biomarkers of early-stage melanoma. Autophagy and beclin 1 regulator 1 (AMBRA1) is a proautophagy regulatory protein with known roles in cell proliferation and differentiation, and is a known tumour suppressor. Loricrin is a marker of epidermal terminal differentiation. What does this study add? AMBRA1 has a functional role in keratinocyte/epidermal proliferation and differentiation. The combined decrease/loss of peritumoral AMBRA1 and loricrin is associated with a significantly increased risk of metastatic spread in American Joint Committee on Cancer (AJCC) stage I tumours vs. melanomas, in which peritumoral AMBRA1 and loricrin are maintained, independently of Breslow depth. What is the translational message? The integration of peritumoral epidermal AMBRA1/loricrin biomarker expression into melanoma care guidelines will facilitate more accurate, personalized risk stratification for patients with AJCC stage I melanomas, thereby facilitating stratification for appropriate follow-up and informing postdiagnostic investigations, including sentinel lymph node biopsy, ultimately resulting in improved disease outcomes and rationalization of healthcare costs.
Collapse
Affiliation(s)
- R Ellis
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,Department of Dermatology, James Cook University Hospital, Middlesbrough, U.K
| | - D Tang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,Department of Dermatology, James Cook University Hospital, Middlesbrough, U.K
| | - B Nasr
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,Department of Pathology, University of North Durham Hospital, Durham, U.K
| | - A Greenwood
- Department of Pathology, James Cook University Hospital, Middlesbrough, U.K
| | - A McConnell
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - M E Anagnostou
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - M Elias
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - S Verykiou
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - D Bajwa
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - T Ewen
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - N J Reynolds
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - P Barrett
- Department of Pathology, University of North Durham Hospital, Durham, U.K
| | - E Carling
- Department of Pathology, St James's University Hospital, Leeds, U.K
| | - G Watson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,Department of Pathology, James Cook University Hospital, Middlesbrough, U.K
| | - J Armstrong
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, U.K
| | - A J Allen
- NIHR Newcastle In Vitro Diagnostics Co-operative, Newcastle University, Newcastle upon Tyne, U.K
| | - S Horswell
- Bioinformatics and Bio Statistics Group, The Francis Crick Institute, London, U.K
| | - M Labus
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - P E Lovat
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| |
Collapse
|
1365
|
Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, Schuebbe G, Renz BW, D’Haese JG, Schloesser H, Heinemann V, Subklewe M, Boeck S, Werner J, von Bergwelt-Baildon M. Advances in cancer immunotherapy 2019 - latest trends. J Exp Clin Cancer Res 2019; 38:268. [PMID: 31217020 PMCID: PMC6585101 DOI: 10.1186/s13046-019-1266-0] [Citation(s) in RCA: 387] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has become an established pillar of cancer treatment improving the prognosis of many patients with a broad variety of hematological and solid malignancies. The two main drivers behind this success are checkpoint inhibitors (CPIs) and chimeric antigen receptor (CAR) T cells. This review summarizes seminal findings from clinical and translational studies recently presented or published at important meetings or in top-tier journals, respectively. For checkpoint blockade, current studies focus on combinational approaches, perioperative use, new tumor entities, response prediction, toxicity management and use in special patient populations. Regarding cellular immunotherapy, recent studies confirmed safety and efficacy of CAR T cells in larger cohorts of patients with acute lymphoblastic leukemia or diffuse large B cell lymphoma. Different strategies to translate the striking success of CAR T cells in B cell malignancies to other hematological and solid cancer types are currently under clinical investigation. Regarding the regional distribution of registered clinical immunotherapy trials a shift from PD-1 / PD-L1 trials (mainly performed in the US and Europe) to CAR T cell trials (majority of trials performed in the US and China) can be noted.
Collapse
Affiliation(s)
- Stephan Kruger
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Bruno L. Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany
| | | | - Gesa Schuebbe
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
| | - Bernhard W. Renz
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan G. D’Haese
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | - Volker Heinemann
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Gene Center LMU, Munich, Germany
| | - Stefan Boeck
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital Munich, LMU Munich, Marchioninistr. 15, D-81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Gene Center LMU, Munich, Germany
| |
Collapse
|
1366
|
A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell 2019; 175:313-326. [PMID: 30290139 DOI: 10.1016/j.cell.2018.09.035] [Citation(s) in RCA: 972] [Impact Index Per Article: 162.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/09/2018] [Accepted: 09/17/2018] [Indexed: 12/28/2022]
Abstract
Harnessing an antitumor immune response has been a fundamental strategy in cancer immunotherapy. For over a century, efforts have primarily focused on amplifying immune activation mechanisms that are employed by humans to eliminate invaders such as viruses and bacteria. This "immune enhancement" strategy often results in rare objective responses and frequent immune-related adverse events (irAEs). However, in the last decade, cancer immunotherapies targeting the B7-H1/PD-1 pathway (anti-PD therapy), have achieved higher objective response rates in patients with much fewer irAEs. This more beneficial tumor response-to-toxicity profile stems from distinct mechanisms of action that restore tumor-induced immune deficiency selectively in the tumor microenvironment, here termed "immune normalization," which has led to its FDA approval in more than 10 cancer indications and facilitated its combination with different therapies. In this article, we wish to highlight the principles of immune normalization and learn from it, with the ultimate goal to guide better designs for future cancer immunotherapies.
Collapse
|
1367
|
Abstract
The prognosis of patients with metastatic melanoma has dramatically improved in recent years with the introduction of two new therapeutic strategies. BRAF and MEK inhibitors are small molecules that are able to block the mitogen-activated protein kinase (MAPK) pathway, which is constitutively activated by recurrent BRAF V600 mutations in 45% of melanoma patients. These agents were shown to provide a rapid and strong response but are often limited by a high rate of secondary resistance. Monoclonal antibodies against the immune checkpoints cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) can restore an efficient and durable anti-tumor immunity, even following treatment discontinuation. Anti-PD-1 antibodies were shown to prolong survival of metastatic melanoma patients and a real cure seems to be obtainable in some patients. Many more therapies are currently under investigation, given that 50% of patients still do not have long-term benefits from approved treatments. The main goal is to avoid or circumvent primary or secondary immune resistance to anti-PD-1 therapy not only by targeting other players in the tumor microenvironment but also by optimizing treatment sequencing and combining anti-PD-1 with other treatments, especially with BRAF and MEK inhibitors. The unexpected major successes of immunotherapies in melanoma have opened the way for the development of these treatments in other cancers. In this review, we describe the different available treatments, their toxicities, and the key components of our decisional algorithms, and give an overview of what we expect to be the near future of melanoma treatment.
Collapse
Affiliation(s)
- Nausicaa Malissen
- Dermatology and Skin Cancer Department, Aix-Marseille University, 264, rue Saint-Pierre, 13385, Marseille, France
| | - Jean-Jacques Grob
- Dermatology and Skin Cancer Department, Aix-Marseille University, 264, rue Saint-Pierre, 13385, Marseille, France.
| |
Collapse
|
1368
|
Abstract
PURPOSE OF REVIEW Novel immunotherapies such as checkpoint inhibitors, bispecific antibodies, and chimeric antigen receptor T cells are leading to promising responses when treating solid tumors and hematological malignancies. T cell neoplasms include leukemia and lymphomas that are derived from T cells and overall are characterized by poor clinical outcomes. This review describes the rational and preliminary results of immunotherapy for patients with T cell lymphoma and leukemia. RECENT FINDINGS For T cell neoplasms, despite significant research effort, only few agents, such as monoclonal antibodies and allogeneic stem cell transplantation, showed some clinical activity. One of the major hurdles to targeting T cell neoplasms is that activation or elimination of T cells, either normal or neoplastic, can cause significant toxicity. A need to develop novel safe and effective immunotherapies for T cell neoplasms exists. In this review, we will discuss the rationale for immunotherapy of T cell leukemia and lymphoma and present the most recent therapeutic approaches.
Collapse
|
1369
|
Elias M, Behbahani S, Maddukuri S, John A, Schwartz R, Lambert W. Prolonged overall survival following metastasectomy in stage
IV
melanoma. J Eur Acad Dermatol Venereol 2019; 33:1719-1725. [DOI: 10.1111/jdv.15667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022]
Affiliation(s)
- M.L. Elias
- Rutgers New Jersey Medical School Newark NJ USA
| | | | | | - A.M. John
- Rutgers Robert Wood Johnson Medical School Piscataway Township NJ USA
| | | | - W.C. Lambert
- Rutgers New Jersey Medical School Newark NJ USA
- Department of Pathology, Immunology, and Laboratory Medicine and of Dermatology Rutgers New Jersey Medical School Newark NJ USA
| |
Collapse
|
1370
|
Kennedy LC, Bhatia S, Thompson JA, Grivas P. Preexisting Autoimmune Disease: Implications for Immune Checkpoint Inhibitor Therapy in Solid Tumors. J Natl Compr Canc Netw 2019; 17:750-757. [DOI: 10.6004/jnccn.2019.7310] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022]
Abstract
The use of immune checkpoint inhibitors (ICIs) is rapidly expanding to the treatment of many cancer types, both in the metastatic setting and as an adjuvant to other therapies. Clinical trials using ICIs have largely excluded patients with preexisting autoimmune diseases due to concerns for increased toxicity. However, emerging evidence shows that ICIs may be considered in some patients with autoimmunity. This review discusses the commonalities between clinical autoimmune diseases and ICI-induced immunotherapy-related adverse events, and summarizes the existing case series that describes patients with solid tumors who have a preexisting autoimmune disease. This review also discusses which patients with autoimmunity could be considered reasonable candidates for ICI therapy.
Collapse
Affiliation(s)
- Laura C. Kennedy
- aDepartment of Medicine, Division of Oncology, University of Washington, and
- bClinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Shailender Bhatia
- aDepartment of Medicine, Division of Oncology, University of Washington, and
- bClinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - John A. Thompson
- aDepartment of Medicine, Division of Oncology, University of Washington, and
- bClinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Petros Grivas
- aDepartment of Medicine, Division of Oncology, University of Washington, and
- bClinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
1371
|
Pavri SN, Han G, Khan S, Han D. Does sentinel lymph node status have prognostic significance in patients with acral lentiginous melanoma? J Surg Oncol 2019; 119:1060-1069. [PMID: 30883783 DOI: 10.1002/jso.25445] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prognostic benefit of sentinel lymph node biopsy (SLNB) and factors predictive of survival specifically in patients with acral lentiginous melanoma (ALM) are unknown. METHODS The SEER database was queried for ALM cases that underwent SLNB from 1998 to 2013. Clinicopathological factors were correlated with SLN status, overall survival (OS), and melanoma-specific survival (MSS). RESULTS Median age for the 753 ALM study patients was 65 years, and 48.2% were male. Median thickness was 2 mm with 38.1% of cases having ulceration. SLN metastases were detected in 194 of 753 cases (25.7%). Multivariable analysis showed that thickness, Clark level IV-V, and ulceration significantly predicted for SLN metastasis (P < 0.05). For patients with positive SLN, 5-year OS and MSS were significantly worse at 48.1% and 58.9%, respectively, compared with 78.7% and 88.5%, respectively, for patients with negative SLN (P < 0.0001). On multivariable analyses, older age, male gender, increasing thickness, ulceration, and a positive SLN significantly predicted for worse OS and MSS (all P < 0.05). CONCLUSION This study confirms the important role of SLNB in ALM. SLN metastases are seen in 25.7% of ALM cases, providing significant prognostic information. In addition, thickness, ulceration status, and SLNB status significantly predict survival in patients with ALM.
Collapse
Affiliation(s)
- Sabrina N Pavri
- Aesthetic and Reconstructive Surgery Institute, UF Health Cancer Center-Orlando Health, Orlando, Florida
| | - Gang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, Texas
| | - Sajid Khan
- Section of Surgical Oncology, Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Dale Han
- Division of Surgical Oncology, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
1372
|
Brokāne L, Jaunalksne I, Tilgase A, Olmane E, Petroška D, Rasa A, Alberts P. Combination treatment with nivolumab and Rigvir of a progressive stage IIC skin melanoma patient. Clin Case Rep 2019; 7:1191-1196. [PMID: 31183092 PMCID: PMC6552946 DOI: 10.1002/ccr3.2182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/01/2019] [Accepted: 04/16/2019] [Indexed: 11/30/2022] Open
Abstract
A 35-year-old male patient was diagnosed with stage IIC skin melanoma that rapidly progressed after surgery. Treatment was continued with radiotherapy, which did not stop further spread of disease and the patient was put on a combination of nivolumab and Rigvir. Subsequently, the progression has slowed.
Collapse
Affiliation(s)
| | | | | | - Evija Olmane
- Department of RadiologyPauls Stradiņš Clinical University HospitalRīgaLatvia
| | - Donatas Petroška
- National Center of PathologyAffiliate of Vilnius University Hospital Santaros KlinikosVilniusLithuania
| | | | | |
Collapse
|
1373
|
Mason R, Au L, Ingles Garces A, Larkin J. Current and emerging systemic therapies for cutaneous metastatic melanoma. Expert Opin Pharmacother 2019; 20:1135-1152. [PMID: 31025594 DOI: 10.1080/14656566.2019.1601700] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Melanoma therapies have evolved rapidly, and initial successes have translated into survival gains for patients with advanced melanoma. Both targeted and immune-therapy now have evidence in earlier stage disease. There are many new agents and combinations of treatments in development as potential future treatment options. This highlights the need for a reflection on current treatment practice trends that are guiding the development of potential new therapies. AREAS COVERED In this review, the authors discuss the evidence for currently approved therapies for cutaneous melanoma, including adjuvant therapy, potential new biomarkers, and emerging treatments with early phase clinical trial data. The authors have searched both the PubMed and clinicaltrials.gov databases for published clinical trials and discuss selected landmark trials of current therapies and of investigational treatment strategies with early evidence for the treatment of melanoma. EXPERT OPINION Significant efficacy has been demonstrated with both immune checkpoint inhibitors and targeted therapies in treating advanced melanoma. A multitude of novel therapies are in development and there is need for instructive biomarker assessment to identify patients likely to respond or be refractory to current therapies, to identify mechanisms of resistance and to direct further treatment options to patients based on individual disease biology.
Collapse
Affiliation(s)
- Robert Mason
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- b Department of Medical Oncology , Gold Coast University Hospital , Southport , Queensland , Australia
| | - Lewis Au
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
| | - Alvaro Ingles Garces
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
| | - James Larkin
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
- d Consultant Oncologist , The Royal Marsden Hospital , London , UK
| |
Collapse
|
1374
|
Hawkins ML, Rioth MJ, Eguchi MM, Cockburn M. Poor prognosis for thin ulcerated melanomas and implications for a more aggressive approach to treatment. J Am Acad Dermatol 2019; 80:1640-1649. [PMID: 30654077 PMCID: PMC7232854 DOI: 10.1016/j.jaad.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/26/2018] [Accepted: 01/06/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Clinical guidelines for the treatment of melanoma are based largely on the behavior of thicker tumors. As a result, little is known about survival differences among patients with thinner tumors. OBJECTIVE To investigate the variability in survival for American Joint Committee on Cancer stage T1 thin melanoma tumors, defined as tumors less than 1 mm thick at diagnosis. METHODS This population-based series included 43,008 non-Hispanic whites in whom cutaneous melanoma was diagnosed between 2004 and 2013 from the California Cancer Registry. Survival outcomes were estimated using the Kaplan-Meier method. Cox proportional hazard models were used to estimate risk of death. RESULTS Survival for patients with thin ulcerated tumors was comparable to that for patients with stage II tumors, who are currently treated more aggressively. At 12 months, patients with thin ulcerated tumors had approximately 6% lower survival (92.5% [95% confidence interval (CI), 90.6%-93.9%]) compared with patients with thin nonulcerated tumors (98.2% [95% CI, 98.0%-98.3%]). At 24 months, this survival difference increased (85.2% [95% CI, 82.8%-87.4%] vs 96.1% [95% CI, 95.8-96.3%] for those with thin ulcerated and thin nonulcerated tumors, respectively) and a greater than 15% survival difference was seen at 60 months. LIMITATIONS Previous reports of cancer registry data have noted some evidence of miscoding of thin tumors. CONCLUSION The poorer survival in patients with ulcerated tumors less than 1 mm thick implies the need for additional studies to determine potential benefits of more aggressive treatment.
Collapse
Affiliation(s)
- Makenzie L Hawkins
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| | - Matthew J Rioth
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado; Department of Medicine-Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| | - Megan M Eguchi
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| | - Myles Cockburn
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado; Department of Dermatology, School of Medicine, University of Colorado, Aurora, Colorado; Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California; Department of Dermatology, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
1375
|
Immune Checkpoint Inhibitors and Beyond: An Overview of Immune-Based Therapies in Merkel Cell Carcinoma. Am J Clin Dermatol 2019; 20:391-407. [PMID: 30784027 DOI: 10.1007/s40257-019-00427-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin cancer. Until 2017, patients with advanced disease were typically treated with conventional chemotherapies, with a median response duration of 3 months. Increased evidence of the role of the immune system in controlling this cancer has paved the way for immune-based therapies, with programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) inhibitors at the frontline. Avelumab, an anti-PD-L1 antibody, was the first-ever drug approved in advanced MCC after showing meaningful efficacy in a second-line setting. Objective responses were observed in one-third of patients and, most importantly, were durable with half of patients and one-third of patients still alive at 1 and 2 years, respectively. When used in a first-line setting, PD-1/PD-L1 inhibitors (avelumab, pembrolizumab, nivolumab) are even more promising as objective responses are observed in approximately 50-70% of patients within the first 4-8 weeks of treatment. Safety profiles are acceptable with 10-20% of patients experiencing adverse events grade ≥ 3. PD-1/PD-L1 inhibitors are considered the standard of care in advanced MCC and are currently being investigated in the adjuvant and neoadjuvant settings. However, innovative treatments are still needed in the metastatic setting, as approximately 50% of these patients will not persistently respond to currently available immunotherapies, and no predictors of response are available yet. Therefore, other immunotherapeutic strategies are now being investigated-ideally in combinations-to enhance the various aspects of the immune response against tumoral cells.
Collapse
|
1376
|
Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, Scolyer RA, Krijgsman O, Sikorska K, Eriksson H, Broeks A, van Thienen JV, Guminski AD, Acosta AT, Ter Meulen S, Koenen AM, Bosch LJW, Shannon K, Pronk LM, Gonzalez M, Ch'ng S, Grijpink-Ongering LG, Stretch J, Heijmink S, van Tinteren H, Haanen JBAG, Nieweg OE, Klop WMC, Zuur CL, Saw RPM, van Houdt WJ, Peeper DS, Spillane AJ, Hansson J, Schumacher TN, Long GV, Blank CU. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol 2019; 20:948-960. [PMID: 31160251 DOI: 10.1016/s1470-2045(19)30151-2] [Citation(s) in RCA: 361] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The outcome of patients with macroscopic stage III melanoma is poor. Neoadjuvant treatment with ipilimumab plus nivolumab at the standard dosing schedule induced pathological responses in a high proportion of patients in two small independent early-phase trials, and no patients with a pathological response have relapsed after a median follow up of 32 months. However, toxicity of the standard ipilimumab plus nivolumab dosing schedule was high, preventing its broader clinical use. The aim of the OpACIN-neo trial was to identify a dosing schedule of ipilimumab plus nivolumab that is less toxic but equally effective. METHODS OpACIN-neo is a multicentre, open-label, phase 2, randomised, controlled trial. Eligible patients were aged at least 18 years, had a WHO performance status of 0-1, had resectable stage III melanoma involving lymph nodes only, and measurable disease according to the Response Evaluation Criteria in Solid Tumors version 1.1. Patients were enrolled from three medical centres in Australia, Sweden, and the Netherlands, and were randomly assigned (1:1:1), stratified by site, to one of three neoadjuvant dosing schedules: group A, two cycles of ipilimumab 3 mg/kg plus nivolumab 1 mg/kg once every 3 weeks intravenously; group B, two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg once every 3 weeks intravenously; or group C, two cycles of ipilimumab 3 mg/kg once every 3 weeks directly followed by two cycles of nivolumab 3 mg/kg once every 2 weeks intravenously. The investigators, site staff, and patients were aware of the treatment assignment during the study participation. Pathologists were masked to treatment allocation and all other data. The primary endpoints were the proportion of patients with grade 3-4 immune-related toxicity within the first 12 weeks and the proportion of patients achieving a radiological objective response and pathological response at 6 weeks. Analyses were done in all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, number NCT02977052, and is ongoing with an additional extension cohort and to complete survival analysis. FINDINGS Between Nov 24, 2016 and June 28, 2018, 105 patients were screened for eligibility, of whom 89 (85%) eligible patients were enrolled and randomly assigned to one of the three groups. Three patients were excluded after randomisation because they were found to be ineligible, and 86 received at least one dose of study drug; 30 patients in group A, 30 in group B, and 26 in group C (accrual to this group was closed early upon advice of the Data Safety Monitoring Board on June 4, 2018 because of severe adverse events). Within the first 12 weeks, grade 3-4 immune-related adverse events were observed in 12 (40%) of 30 patients in group A, six (20%) of 30 in group B, and 13 (50%) of 26 in group C. The difference in grade 3-4 toxicity between group B and A was -20% (95% CI -46 to 6; p=0·158) and between group C and group A was 10% (-20 to 40; p=0·591). The most common grade 3-4 adverse events were elevated liver enzymes in group A (six [20%)]) and colitis in group C (five [19%]); in group B, none of the grade 3-4 adverse events were seen in more than one patient. One patient (in group A) died 9·5 months after the start of treatment due to the consequences of late-onset immune-related encephalitis, which was possibly treatment-related. 19 (63% [95% CI 44-80]) of 30 patients in group A, 17 (57% [37-75]) of 30 in group B, and nine (35% [17-56]) of 26 in group C achieved a radiological objective response, while pathological responses occurred in 24 (80% [61-92]) patients in group A, 23 (77% [58-90]) in group B, and 17 (65% [44-83]) in group C. INTERPRETATION OpACIN-neo identified a tolerable neoadjuvant dosing schedule (group B: two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg) that induces a pathological response in a high proportion of patients and might be suitable for broader clinical use. When more mature data confirm these early observations, this schedule should be tested in randomised phase 3 studies versus adjuvant therapies, which are the current standard-of-care systemic therapy for patients with stage III melanoma. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
| | - Alexander M Menzies
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | - Chandra Adhikari
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Richard A Scolyer
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Hanna Eriksson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | | | - Alexander D Guminski
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Kerwin Shannon
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Loes M Pronk
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria Gonzalez
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Sydney Ch'ng
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | - Jonathan Stretch
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Stijn Heijmink
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Omgo E Nieweg
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Robyn P M Saw
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Andrew J Spillane
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Johan Hansson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | - Georgina V Long
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
1377
|
Dillman RO, Cornforth AN, McClay EF, Depriest C. Patient-specific dendritic cell vaccines with autologous tumor antigens in 72 patients with metastatic melanoma. Melanoma Manag 2019; 6:MMT20. [PMID: 31406564 PMCID: PMC6688559 DOI: 10.2217/mmt-2018-0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Metastatic melanoma patients were treated with patient-specific vaccines consisting of autologous dendritic cells loaded with antigens from irradiated cells from short-term autologous tumor cell lines. Patients & methods: A total of 72 patients were enrolled in a single-arm Phase I/II (NCT00948480) trial or a randomized Phase II (NCT00436930). Results: Toxicity was minimal. Median overall survival (OS) was 49.4 months; 5-year OS 46%. A 5-year OS was 72% for 18 recurrent stage 3 without measurable disease when treated and 53% for 30 stage 4 without measurable disease when treated. A total of 24 patients with measurable stage 4 when treated (median of four prior therapies) had an 18.5 months median OS and 46% 2-year OS. Conclusion: This dendritic cell vaccine was associated with encouraging survival in all three clinical subsets. Clinicaltrial.gov NCT00436930 and NCT00948480.
Collapse
Affiliation(s)
- Robert O Dillman
- AIVITA Biomedical, Inc., Irvine, CA 92612, USA.,Hoag Cancer Institute, Newport Beach, CA 92658, USA.,University of California, Irvine, CA 92697, USA
| | | | - Edward F McClay
- California Cancer Associates for Research & Excellence (cCARE), Institute for Melanoma Research & Education, Encinitas, CA 92024, USA
| | | |
Collapse
|
1378
|
Nakamura Y. Biomarkers for Immune Checkpoint Inhibitor-Mediated Tumor Response and Adverse Events. Front Med (Lausanne) 2019; 6:119. [PMID: 31192215 PMCID: PMC6549005 DOI: 10.3389/fmed.2019.00119] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
In the last decade, inhibitors targeting immune checkpoint molecules such as cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 (PD-1), and programmed cell death-ligand 1 (PD-L1) brought about a major paradigm shift in cancer treatment. These immune checkpoint inhibitors (ICIs) improved the overall survival of a variety of cancer such as malignant melanoma and non-small lung cancer. In addition, numerous clinical trials for additional indication of ICIs including adjuvant and neo-adjuvant therapies are also currently ongoing. Therefore, more and more patients will receive ICIs in the future. However, despite the improved outcome of the cancer treatment by ICIs, the efficacy remains still limited and tumor regression have not been obtained in many cancer patients. In addition, treatment with ICIs is also associated with substantial toxicities, described as immune-related adverse events (irAEs). Therefore, biomarkers to predict tumor response and occurrence of irAEs by the treatment with ICIs are required to avoid overtreatment of ICIs and minimize irAEs development. Whereas, numerous factors have been reported as potential biomarkers for tumor response to ICIs, factors for predicting irAE have been less reported. In this review, we show recent advances in the understanding of biomarkers for tumor response and occurrence of irAEs in cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
1379
|
[Reprint of: New guidelines for stage III melanoma (the French Cutaneous Oncology Group)]. Bull Cancer 2019; 106:560-573. [PMID: 31122657 DOI: 10.1016/j.bulcan.2019.04.001] [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: 10/31/2018] [Accepted: 01/15/2019] [Indexed: 11/22/2022]
Abstract
Improved knowledge of sentinel node procedures coupled with the results of adjuvant clinical trials in stage III melanoma have prompted the French Cutaneous Oncology Group to propose new guidelines for the management of stage III melanoma. These guidelines comply with the principles of the evidence-based medicine.
Collapse
|
1380
|
Bernicker EH. What the Oncologist Needs From the Pathologist for Tyrosine Kinase Inhibitor Therapies. Arch Pathol Lab Med 2019; 143:1089-1092. [PMID: 31100016 DOI: 10.5858/arpa.2019-0210-sa] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Eric H Bernicker
- From Thoracic and Uveal Melanoma Medical Oncology, Cancer Center, Houston Methodist Hospital, Houston, Texas; and Clinical Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
1381
|
Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, Liu X. Application of PD-1 Blockade in Cancer Immunotherapy. Comput Struct Biotechnol J 2019; 17:661-674. [PMID: 31205619 PMCID: PMC6558092 DOI: 10.1016/j.csbj.2019.03.006] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023] Open
Abstract
The programmed cell death protein 1 (PD-1) pathway has received considerable attention due to its role in eliciting the immune checkpoint response of T cells, resulting in tumor cells capable of evading immune surveillance and being highly refractory to conventional chemotherapy. Application of anti-PD-1/PD-L1 antibodies as checkpoint inhibitors is rapidly becoming a promising therapeutic approach in treating tumors, and some of them have successfully been commercialized in the past few years. However, not all patients show complete responses and adverse events have been noted, suggesting a better understanding of PD-1 pathway mediated immunosuppression is needed to predict patient response and improve treatment efficacy. Here, we review the progresses on the studies of the mechanistic role of PD-1 pathway in the tumor immune evasion, recent clinical development and commercialization of PD-1 pathway inhibitors, the toxicities associated with PD-1 blockade observed in clinical trials as well as how to improve therapeutic efficacy and safety of cancer immunotherapy.
Collapse
Key Words
- 5-AZA-dC, 5-aza-2′-deoxycytidine
- ADCC, Antibody-dependent cellular cytotoxicity
- AEs, Adverse events
- AP1, Activator protein 1
- APCs, Antigen presenting cells
- ASCT, Autologous stem cell transplantation
- B2M, β2 microglobulin
- BATF, Basic leucine zipper transcriptional factor ATF-like
- BICR, Blinded Independent Central Review
- BV, Brentuximab vedotin
- CC, Cervical cancer
- CRC, Colorectal cancer
- CTLA-4, Cytotoxic T-lymphocyte–associated antigen 4
- CXCL9, C-X-C motif chemokine ligand 9
- Checkpoint blockade
- DCM, Dilated cardiomyopathy
- DCs, Dendritic cells
- DNMT, DNA methyltransferase
- DOR, Duration overall response
- DZNep, 3-Deazaneplanocin A
- ERK, Extracellular signal–regulated kinase
- EZH2, Enhancer of zeste homolog 2
- GC, Gastric cancer
- GEJ, GASTRIC or gastroesophageal junction
- HCC, Hepatocellular carcinoma
- HNSCC, Head and neck squamous cell carcinoma
- HR, Hazard ratio
- ICC, Investigator-choice chemotherapy
- ICOS, Inducible T-cell co-stimulator
- IFN, Interferon
- IHC, Immunohistochemistry
- ITIM, Immune-receptortyrosine-based inhibitory motif
- ITSM, Immune-receptortyrosine-based switch motif
- ITT, Intention-to-treat
- Immune surveillance
- Immunotherapy
- IrAEs, Immune related adverse events
- JMJD3, Jumonji Domain-Containing Protein 3
- LAG3, Lymphocyte-activation gene 3
- LCK, Tyrosine-protein kinase Lck
- MAP, Mitogen-activated protein
- MCC, Merkel cell carcinoma
- MHC, Major histocompatibility
- MSI-H, Microsatellite instability-high
- NF-κB, Nuclear factor-κB
- NFAT, Nuclear factor of activated T cells
- NSCLC, Non-small cell lung cancer
- ORR, Overall response rate
- OS, Overall survival
- PD-1
- PD-1, Programmed cell death 1
- PD-L1
- PD-L1, Programmed death-ligand 1
- PFS, Progression-free survival
- PI3K, Phosphoinositide 3-kinase
- PKC, Protein kinase C
- PMBCL, Primary mediastinal large B-cell lymphoma
- PRC2, Polycomb repressive complex 2
- PTEN, Phosphatase and tensin homolog
- PTPs, Protein tyrosine phosphatases
- RCC, Renal cell carcinoma
- SCLC, Small cell lung cancer
- SHP2, Src homology 2 domain-containing phosphatase 2
- SIRPα, Signal-regulatory protein alpha
- TCR, T-cell receptor
- TGF, Transforming growth factor
- TIICs, Tumor infiltrating immune cells
- TILs, Tumor-infiltrating lymphocytes
- TIM3, T-cell immunoglobulin and mucin-domain containing-3
- TMB, Tumor mutation burden
- TME, Tumor microenvironment
- UC, Urothelial carcinoma
- VEGF, Vascular endothelial growth factor
- ZAP70, Zeta-chain-associated protein kinase 70
- cHL, Classical Hodgkin lymphoma
- cTnI, Cardiac troponin I
- dMMR, DNA mismatch repair deficiency
Collapse
Affiliation(s)
- Xiaomo Wu
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
- Department of Biomedicine, University of Basel, Klingelbergstr. 70, CH-4056 Basel, Switzerland
| | - Zhongkai Gu
- The Institute of Biomedical Sciences, Fudan University, Mingdao Building, Dongan Road 131, Shanghai 200032, PR China
| | - Yang Chen
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Borui Chen
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Wei Chen
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Chazhong Road 20, Fuzhou 350005, PR China
| | - Liqiang Weng
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou 350025, PR China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou 350025, PR China
| |
Collapse
|
1382
|
Cabel L, Proudhon C, Romano E, Girard N, Lantz O, Stern MH, Pierga JY, Bidard FC. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat Rev Clin Oncol 2019; 15:639-650. [PMID: 30050094 DOI: 10.1038/s41571-018-0074-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Considerable interest surrounds the use of immune-checkpoint inhibitors in patients with solid tumours following the demonstration of the impressive clinical efficacy of anti-programmed cell death protein 1 and anti-programmed cell death 1 ligand 1 antibodies in several tumour types. However, the emergence of unexpected tumour response patterns, such as pseudoprogression or hyperprogression, might complicate the management of patients receiving these agents. Analysis of circulating tumour DNA (ctDNA) has been shown to have prognostic value by enabling the detection of residual proliferating disease in the adjuvant setting and estimation of tumour burden in the metastatic setting, which are key stratification biomarkers for use of immune-checkpoint inhibition (ICI). Furthermore, examinations of ctDNA for genetic predictors of responsiveness to immunotherapy, such as mutations, tumour mutational load, and microsatellite instability provide a noninvasive surrogate for tumour biopsy sampling. Proof-of-concept reports have also demonstrated that quantitative changes in ctDNA levels early in the course of disease are a promising tool for the assessment of responsiveness to ICI that might complement standard imaging approaches. Other applications of this technology are also currently under investigation, such as early detection of resistance to immunotherapy and characterization of mechanisms of resistance. The aim of this Review is to summarize available data on the application of ctDNA in patients receiving immunotherapy and to discuss the most promising future directions.
Collapse
Affiliation(s)
- Luc Cabel
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France.,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Charlotte Proudhon
- Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France
| | - Emanuela Romano
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Nicolas Girard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Olivier Lantz
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Marc-Henri Stern
- INSERM U830, Institut Curie, PSL Research University, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France.,Paris Descartes University, Paris, France
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France. .,Versailles Saint Quentin en Yvelines University, Paris Saclay University, Saint Cloud, Paris, France. .,Circulating Tumor Biomarkers Laboratory, Institut Curie, PSL Research University, Paris, France.
| |
Collapse
|
1383
|
Postsurgical treatment landscape and economic burden of locoregional and distant recurrence in patients with operable nonmetastatic melanoma. Melanoma Res 2019; 28:618-628. [PMID: 30216199 PMCID: PMC6221390 DOI: 10.1097/cmr.0000000000000507] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Supplemental Digital Content is available in the text. Surgery is the mainstay treatment for operable nonmetastatic melanoma, but recurrences are common and limit patients’ survival. This study aimed to describe real-world patterns of treatment and recurrence in patients with melanoma and to quantify healthcare resource utilization (HRU) and costs associated with episodes of locoregional/distant recurrences. Adults with nonmetastatic melanoma who underwent melanoma lymph node surgery were identified from the Truven Health MarketScan database (1 January 2008 to 31 July 2017). Locoregional and distant recurrence(s) were identified on the basis of postsurgery recurrence indicators (i.e. initiation of new melanoma pharmacotherapy, new radiotherapy, or new surgery; secondary malignancy diagnoses). Of 6400 eligible patients, 219 (3.4%) initiated adjuvant therapy within 3 months of surgery, mostly with interferon α-2b (n=206/219, 94.1%). A total of 1191/6400 (18.6%) patients developed recurrence(s) over a median follow-up of 23.1 months (102/6400, 1.6% distant recurrences). Among the 219 patients initiated on adjuvant therapy, 73 (33.3%) experienced recurrences (distant recurrences: 13/219, 5.9%). The mean total all-cause healthcare cost was $2645 per patient per month (PPPM) during locoregional recurrence episodes and $12 940 PPPM during distant recurrence episodes. In the year after recurrence, HRU was particularly higher in patients with distant recurrence versus recurrence-free matched controls: by 9.2 inpatient admissions, 54.4 inpatient days, 8.8 emergency department admissions, and 185.9 outpatient visits (per 100 person-months), whereas all-cause healthcare costs were higher by $14 953 PPPM. It remains to be determined whether the new generation of adjuvant therapies, such as immune checkpoint inhibitors and targeted agents, will increase the use of adjuvant therapies, and reduce the risk of recurrences and associated HRU/cost.
Collapse
|
1384
|
Hindié E. What Is the Role of Dabrafenib Plus Trametinib Adjuvant Therapy in Stage IIIA Melanoma? J Clin Oncol 2019; 37:1355-1356. [DOI: 10.1200/jco.18.02075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Elif Hindié
- Elif Hindié, MD, PhD, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
1385
|
Vucic S. Immune checkpoint inhibitors and neuropathy: A new dawn. Clin Neurophysiol 2019; 130:1401-1402. [PMID: 31101419 DOI: 10.1016/j.clinph.2019.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Steve Vucic
- Sydney Medical School Westmead, University of Sydney, Australia.
| |
Collapse
|
1386
|
Brooker RC, Sacco JJ, Schache AG. Integration of Checkpoint Inhibitors into the Management of Locally Advanced Head and Neck Cancer - Future Perspectives. Clin Oncol (R Coll Radiol) 2019; 31:424-431. [PMID: 31097366 DOI: 10.1016/j.clon.2019.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/01/2019] [Accepted: 03/20/2019] [Indexed: 10/26/2022]
Affiliation(s)
- R C Brooker
- The Department of Molecular and Clinical Cancer Medicine, The Northwest Cancer Research Centre, University of Liverpool, Liverpool, UK; The Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, UK.
| | - J J Sacco
- The Department of Molecular and Clinical Cancer Medicine, The Northwest Cancer Research Centre, University of Liverpool, Liverpool, UK; The Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, UK
| | - A G Schache
- The Department of Molecular and Clinical Cancer Medicine, The Northwest Cancer Research Centre, University of Liverpool, Liverpool, UK; Head and Neck Unit, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
1387
|
Yushak M, Mehnert J, Luke J, Poklepovic A. Approaches to High-Risk Resected Stage II and III Melanoma. Am Soc Clin Oncol Educ Book 2019; 39:e207-e211. [PMID: 31099653 DOI: 10.1200/edbk_239283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Over the last decade, several therapies, including both targeted and immune checkpoint inhibitors, have dramatically changed the treatment landscape for patients with metastatic melanoma. These same therapies are now being used in the adjuvant setting with the hope of delaying or preventing the development of metastatic disease. Although phase III trials have shown a clear benefit for patients with resected bulky nodal disease, treatment decisions for patients with earlier-stage (high-risk stage II and stage IIIA) melanoma in the adjuvant setting are less straightforward given the small number of patients studied so far. Among patients with stage IIIB and worse disease, both targeted and immune checkpoint inhibitors have shown benefit in recurrence-free survival. Although a head-to-head comparison has not been completed, patient and tumor characteristics can guide the optimal treatment of an individual.
Collapse
Affiliation(s)
- Melinda Yushak
- 1 Winship Cancer Institute, Emory University, Atlanta, GA
| | - Janice Mehnert
- 2 Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Jason Luke
- 3 University of Chicago Medicine, Chicago, IL
| | - Andrew Poklepovic
- 4 Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
1388
|
Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front Oncol 2019; 9:396. [PMID: 31139574 PMCID: PMC6527887 DOI: 10.3389/fonc.2019.00396] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
There are a wide range of therapies for metastatic colorectal cancer (CRC) available, but outcomes remain suboptimal. Learning the role of the immune system in cancer development and progression led to advances in the treatment over the last decade. While the field is rapidly evolving, PD-1, and PD-L1 inhibitors have a leading role amongst immunomodulatory agents. They act against pathways involved in adaptive immune suppression resulting in immune checkpoint blockade. Immunotherapy has been slow to impact the management of this patient population due to disappointing results, mainly when used broadly. Nevertheless, some patients with microsatellite-instability-high (MSI-H) or mismatch repair-deficient (dMMR) CRC appear to be susceptible to checkpoint inhibitors with objective and sustained clinical responses, providing a new therapeutic option for patients with advanced disease. This article provides a comprehensive review of the early and late phase trials with the updated data of PD-1/PD-L1 inhibitors alone or in combination with other therapies (immunotherapy, targeted therapy and chemotherapy). While data is still limited, many ongoing trials are underway, testing the efficacy of these agents in CRC. Current and future challenges of PD-1 and PD-L1 inhibitors are also discussed.
Collapse
Affiliation(s)
- André F. Oliveira
- Serviço de Oncologia Médica, Centro Hospitalar Universitário Algarve, Faro, Portugal
| | | | | |
Collapse
|
1389
|
Audrito V, Managò A, Gaudino F, Deaglio S. Targeting metabolic reprogramming in metastatic melanoma: The key role of nicotinamide phosphoribosyltransferase (NAMPT). Semin Cell Dev Biol 2019; 98:192-201. [PMID: 31059816 DOI: 10.1016/j.semcdb.2019.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022]
Abstract
Cancer cells rewire their metabolism to support proliferation, growth and survival. In metastatic melanoma the BRAF oncogenic pathway is a master regulator of this process, highlighting the importance of metabolic reprogramming in the pathogenesis of this tumor and offering potential therapeutic approaches. Metabolic adaptation of melanoma cells generally requires increased amounts of NAD+, an essential redox cofactor in cellular metabolism and a signaling molecule. Nicotinamide phosphoribosyltransferase (NAMPT) is the most important NAD+ biosynthetic enzyme in mammalian cells and a direct target of the BRAF oncogenic signaling pathway. These findings suggest that NAMPT is an attractive new therapeutic target, particularly in combination strategies with BRAF or MEK inhibitors. Here we review current knowledge on how oncogenic signaling reprograms metabolism in BRAF-mutated melanoma, and discuss how NAMPT/NAD+ axis contributes to these processes. Lastly, we present evidence supporting a role of NAMPT as a novel therapeutic target in metastatic melanoma.
Collapse
Affiliation(s)
- Valentina Audrito
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy.
| | - Antonella Managò
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy
| | - Federica Gaudino
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy; Italian Institute for Genomic Medicine, Turin, Italy.
| |
Collapse
|
1390
|
Ascierto PA, Bruzzi P, Eggermont A, Hamid O, Tawbi HA, van Akkooi A, Testori A, Caracò C, Puzanov I, Perrone F. The great debate at "Melanoma Bridge 2018", Naples, December 1st, 2018. J Transl Med 2019; 17:148. [PMID: 31077205 PMCID: PMC6509811 DOI: 10.1186/s12967-019-1892-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
The great debate session at the 2018 Melanoma Bridge congress (November 29-December 1, Naples, Italy) featured counterpoint views from experts on three topical issues in melanoma. These were whether overall survival should still be the main endpoint for clinical trials in melanoma, whether anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4 is still the optimal choice of drug to use in combination with an anti-programmed death (PD)/PD-ligand (L)-1 agent, and the place of adjuvant versus neoadjuvant therapy in patients with melanoma. These three important debates are summarised 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.
| | - Paolo Bruzzi
- Clinical Epidemiology Unit, University Hospital "San Martino", Genoa, Italy
| | | | - Omid Hamid
- Clinical Research and Immunotherapy, The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | - Hussein A Tawbi
- Melanoma Clinical Research & Early Drug Development, Melanoma Medical Oncology, Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands
| | | | - Corrado Caracò
- Department Melanoma, Soft Tissue, Muscle-Skeletal and Head-Neck, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| |
Collapse
|
1391
|
Marra A, Viale G, Curigliano G. Recent advances in triple negative breast cancer: the immunotherapy era. BMC Med 2019; 17:90. [PMID: 31068190 PMCID: PMC6507064 DOI: 10.1186/s12916-019-1326-5] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Several accomplishments have been achieved in triple-negative breast cancer (TNBC) research over the last year. The phase III IMpassion130 trial comparing chemotherapy plus atezolizumab versus chemotherapy plus placebo brought breast cancer into the immunotherapy era. Nevertheless, despite encouraging results being obtained in this trial, many open questions remain. MAIN BODY A positive overall survival outcome was achieved only in PD-L1+ TNBC patients, suggesting a need to enrich the patient population more likely to benefit from an immunotherapeutic approach. Moreover, it remains unknown whether single-agent immunotherapy might be a good option for some patients. In this context, the discovery and implementation of novel and appropriate biomarkers are required. Focusing on the early onset of TNBC, neoadjuvant trials could represent excellent in vivo platforms to test immunotherapy agents and their potential combinations, allowing the performance of translational studies for biomarker implementation and improved patient selection. CONCLUSION The aim of our review is to present recent advances in TNBC treatment and to discuss open issues in order to better define potential future directions for immunotherapy in TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milano, Milano, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milano, Milano, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy. .,Department of Oncology and Haematology, University of Milano, Milano, Italy.
| |
Collapse
|
1392
|
Martinez Pena GN, Jiang C. Endobronchial, laryngeal and mediastinal melanoma: a rare constellation of metastatic disease. BMJ Case Rep 2019; 12:e228957. [PMID: 31068348 PMCID: PMC6506047 DOI: 10.1136/bcr-2018-228957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2019] [Indexed: 12/17/2022] Open
Abstract
A 45-year-old man presents with acute respiratory failure. Imaging revealed a left mainstem endobronchial mass with subcarinal lymphadenopathy, but no other evidence of a primary tumour. An incidental laryngeal nodule was found during bronchoscopy. Biopsies of this lesion by nasopharyngoscopy and subcarinal lymph nodes via mediastinoscopy were performed. Histopathological and immunohistochemical examination showed evidence of melanoma in both samples. Mutational analysis identified the presence of a BRAFV600E mutation. The patient underwent bronchoscopic ablation of the left mainstem endobronchial tumour with laser therapy followed by initiation of encorafenib and binimetinib combination therapy. The patient remains alive at 4 months after initial presentation of disease. This case adds to the body of literature highlighting the clinical heterogeneity and challenges of the management of metastatic pulmonary melanoma. To the best of our knowledge, this simultaneous constellation of metastasis has not been described before.
Collapse
Affiliation(s)
| | - Chuan Jiang
- Medicine – Pulmonary Medicine, Jamaica Hospital Medical Center, Jamaica, New York, USA
| |
Collapse
|
1393
|
Luke JJ. Comprehensive Clinical Trial Data Summation for BRAF-MEK Inhibition and Checkpoint Immunotherapy in Metastatic Melanoma. Oncologist 2019; 24:e1197-e1211. [PMID: 31064886 PMCID: PMC6853121 DOI: 10.1634/theoncologist.2018-0876] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/22/2019] [Indexed: 12/30/2022] Open
Abstract
This review focuses on checkpoint and BRAF inhibitors, exploring outcomes based on clinical and disease characteristics to identify trends that might inform treatment decisions for the management of melanoma. Background. Immune checkpoint inhibitors, along with BRAF and MEK inhibitors, have dramatically changed the management of and outlook for patients with metastatic melanoma. Analyses of long‐term follow‐up data and subanalyses based on disease characteristics may inform clinical decision making. Methods. Reports of clinical trials in metastatic melanoma published between January 1, 2012, and August 30, 2018, were identified using PubMed (terms: melanoma AND [dabrafenib OR trametinib OR vemurafenib OR cobimetinib OR encorafenib OR ipilimumab OR nivolumab OR pembrolizumab]) and were systematically reviewed. Relevant congress proceedings were also assessed. Efficacy data from key phase III trials were analyzed and trends identified. Results. Substantial improvements in objective response rates, progression‐free survival, and overall survival were documented across 14 identified publications. Subgroup findings supported that patients with lower disease burden derive greater benefit than patients with more advanced disease, limiting the value of disease burden in the clinical decision‐making process. However, these agents consistently conferred benefits despite the presence of poor prognostic features. Several clinically relevant questions remain, including how best to sequence immune checkpoint inhibitors and combination targeted therapy. Conclusion. This research, coupled with ongoing investigations, including those on predictive biomarkers, suggests that the treatment decision‐making process is likely to become more nuanced. Implications for Practice. The management of melanoma has been rapidly advancing with new classes of agents, including immune checkpoint and BRAF inhibitors. With long‐term follow‐up, their impact on response rates and survival outcomes is well documented. Additional findings from subgroup analyses suggest that patients with lower disease burden derive greater benefit, yet both consistently confer benefit in patients with higher disease burden. Currently, there is a paucity of data to guide first‐line treatment selection between immunotherapy and BRAF‐targeted therapy in clinical practice or to estimate their impact when sequenced. Gaining these insights will facilitate a more nuanced management approach.
Collapse
Affiliation(s)
- Jason J Luke
- Department of Medicine, Division of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
1394
|
Lyon AR, Yousaf N, Battisti NML, Moslehi J, Larkin J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol 2019; 19:e447-e458. [PMID: 30191849 DOI: 10.1016/s1470-2045(18)30457-1] [Citation(s) in RCA: 411] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/11/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors are a new class of anticancer therapies that amplify T-cell-mediated immune responses against cancer cells. Immune checkpoint inhibitors have shown important benefits in phase 3 trials, and several agents have been approved for specific malignancies. Although adverse events from immune checkpoint inhibitors are a common occurrence, cardiotoxic effects are uncommon, but are often serious complications with a relatively high mortality. Most cardiotoxic effects appear to be inflammatory in nature. Clinical assessment of a combination of biomarkers, electrocardiography, cardiac imaging, and endomyocardial biopsy can be used to confirm a possible diagnosis. In this Review, we discuss the epidemiology of immune checkpoint inhibitor-mediated cardiotoxic effects, as well as their clinical presentation, subtypes, risk factors, pathophysiology, and clinical management, including the introduction of a new surveillance strategy.
Collapse
Affiliation(s)
- Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, London, UK; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Nadia Yousaf
- Royal Marsden Hospital NHS Foundation Trust, London, UK
| | | | - Javid Moslehi
- Cardiooncology Program, Vanderbilt School of Medicine, Nashville, TN, USA
| | - James Larkin
- Royal Marsden Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
1395
|
Martinez Chanza N, Tripathi A, Harshman LC. Adjuvant Therapy Options in Renal Cell Carcinoma: Where Do We Stand? Curr Treat Options Oncol 2019; 20:44. [DOI: 10.1007/s11864-019-0639-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
1396
|
Taieb J, André T, Auclin E. Refining adjuvant therapy for non-metastatic colon cancer, new standards and perspectives. Cancer Treat Rev 2019; 75:1-11. [DOI: 10.1016/j.ctrv.2019.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
|
1397
|
Tan L, Sandhu S, Lee RJ, Li J, Callahan J, Ftouni S, Dhomen N, Middlehurst P, Wallace A, Raleigh J, Hatzimihalis A, Henderson MA, Shackleton M, Haydon A, Mar V, Gyorki DE, Oudit D, Dawson MA, Hicks RJ, Lorigan P, McArthur GA, Marais R, Wong SQ, Dawson SJ. Prediction and monitoring of relapse in stage III melanoma using circulating tumor DNA. Ann Oncol 2019; 30:804-814. [PMID: 30838379 PMCID: PMC6551451 DOI: 10.1093/annonc/mdz048] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The advent of effective adjuvant therapies for patients with resected melanoma has highlighted the need to stratify patients based on risk of relapse given the cost and toxicities associated with treatment. Here we assessed circulating tumor DNA (ctDNA) to predict and monitor relapse in resected stage III melanoma. PATIENTS AND METHODS Somatic mutations were identified in 99/133 (74%) patients through tumor tissue sequencing. Personalized droplet digital PCR (ddPCR) assays were used to detect known mutations in 315 prospectively collected plasma samples from mutation-positive patients. External validation was performed in a prospective independent cohort (n = 29). RESULTS ctDNA was detected in 37 of 99 (37%) individuals. In 81 patients who did not receive adjuvant therapy, 90% of patients with ctDNA detected at baseline and 100% of patients with ctDNA detected at the postoperative timepoint relapsed at a median follow up of 20 months. ctDNA detection predicted patients at high risk of relapse at baseline [relapse-free survival (RFS) hazard ratio (HR) 2.9; 95% confidence interval (CI) 1.5-5.6; P = 0.002] and postoperatively (HR 10; 95% CI 4.3-24; P < 0.001). ctDNA detection at baseline [HR 2.9; 95% CI 1.3-5.7; P = 0.003 and postoperatively (HR 11; 95% CI 4.3-27; P < 0.001] was also associated with inferior distant metastasis-free survival (DMFS). These findings were validated in the independent cohort. ctDNA detection remained an independent predictor of RFS and DMFS in multivariate analyses after adjustment for disease stage and BRAF mutation status. CONCLUSION Baseline and postoperative ctDNA detection in two independent prospective cohorts identified stage III melanoma patients at highest risk of relapse and has potential to inform adjuvant therapy decisions.
Collapse
Affiliation(s)
- L Tan
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - S Sandhu
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - R J Lee
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester
| | - J Li
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - J Callahan
- Peter MacCallum Cancer Centre, Melbourne
| | - S Ftouni
- Peter MacCallum Cancer Centre, Melbourne
| | - N Dhomen
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester
| | - P Middlehurst
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester
| | - A Wallace
- Genomic Diagnostics Laboratory, Manchester Centre for Genomic Medicine, Manchester, UK
| | - J Raleigh
- Peter MacCallum Cancer Centre, Melbourne
| | | | - M A Henderson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | | | | | - V Mar
- The Alfred Hospital, Melbourne
| | - D E Gyorki
- Peter MacCallum Cancer Centre, Melbourne; Department of Surgery, The University of Melbourne, Melbourne, Australia
| | - D Oudit
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - M A Dawson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, Australia
| | - R J Hicks
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - P Lorigan
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - G A McArthur
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - R Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester
| | - S Q Wong
- Peter MacCallum Cancer Centre, Melbourne
| | - S-J Dawson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
1398
|
Yang W, Li S, Yang Q. Risk of dermatologic and mucosal adverse events associated with PD-1/PD-L1 inhibitors in cancer patients: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2019; 98:e15731. [PMID: 31096532 PMCID: PMC6531085 DOI: 10.1097/md.0000000000015731] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Programmed death 1 protein (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors are promising cancer immunotherapy. Their dermatologic safety profiles are still poorly understood. The purpose of this article is to evaluate the incidence of selected dermatologic and mucosal adverse effects (AEs) and determine the risk of developing these adverse events associated with PD-1/PD-L1 inhibitors, compared with chemotherapy or ipilimumab. METHODS PubMed was searched for eligible studies (up to February 21, 2019). Only phase II and phase III randomized controlled trials (RCTs) compared with chemotherapy or ipilimumab monotherapy were included in this meta-analysis. RESULTS A total 11,465 patients from 18 clinical trials were included in this meta-analysis. Rash and pruritus were the most frequently reported dermatologic AE, with incidence 11.8% and 12.2% respectively. Compared with patients receiving chemotherapy, PD-1/PD-L1 inhibitor treated patients had higher risk of developing rash (RR = 1.84), pruritus (RR = 3.74) and vitiligo (RR = 9.54), and also lower risk in developing mucosal inflammation (RR = 0.26), stomatitis (RR = 0.26), and alopecia (RR = 0.03). Additionally, anti-PD1/PD-L1 drugs had similar risk of developing rash and lower risk of inducing pruritus compared to ipilimumab. In the subgroup analysis, PD-L1 inhibitor demonstrated better safety than PD-1 inhibitor in developing rash, with RR = 1.38 and RR = 2.11, respectively. CONCLUSION Our meta-analysis concluded that anti PD-1/PD-L1 drugs have different dermatological and mucosal safety profile compared to conventional therapy, and differences of dermatological toxicity between PD-1 and PD-L1 inhibitor warrant further investigation.
Collapse
Affiliation(s)
- Wenwei Yang
- Department of Clinical Medicine, Queen Mary College of Nanchang University, Nanchang, Jiangxi
| | - Shuquan Li
- Department of Clinical Medicine, Queen Mary College of Nanchang University, Nanchang, Jiangxi
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong University Affiliated Provincial Hospital, Jinan, Shandong, China
| |
Collapse
|
1399
|
Keller J, Schwartz TL, Lizalek JM, Chang E, Patel AD, Hurley MY, Hsueh EC. Prospective validation of the prognostic 31-gene expression profiling test in primary cutaneous melanoma. Cancer Med 2019; 8:2205-2212. [PMID: 30950242 PMCID: PMC6536922 DOI: 10.1002/cam4.2128] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/27/2019] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Gene expression profiling (GEP) has been integrated into cancer treatment decision-making in multiple neoplasms. We prospectively evaluated the prognostic utility of the 31-GEP test (DecisionDx-Melanoma, Castle Biosciences, Inc) in cutaneous melanoma (CM) patients undergoing sentinel node biopsy (SNB). METHODS One hundred fifty-nine patients (age 26-88) diagnosed with melanoma between 01/2013 and 8/2015 underwent SNB and concurrent GEP testing. GEP results were reported as low-risk Class 1 (subclasses 1A and 1B) or high-risk Class 2 (subclasses 2A and 2B). Statistical analyses were performed with chi-square analysis, t tests, log-rank tests, and Cox proportional hazard models. Recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) were estimated using Kaplan-Meier method. RESULTS Median follow-up was 44.9 months for event-free cases. Median Breslow thickness was 1.4 mm (0.2-15.0 mm). There were 117 Class 1 and 42 Class 2 patients. Gender, age, Breslow thickness, ulceration, SNB positivity, and AJCC stage were significantly associated with GEP classification (P < 0.05 for all). Recurrence and distant metastasis rates were 5% and 1% for Class 1 patients compared with 55% and 36% for Class 2 patients. Sensitivities of Class 2 and SNB for recurrence were 79% and 34%, respectively. Of 10 SNB-positive/Class 2 patients, 9 recurred. By multivariate analysis, only SNB result and GEP class were statistically associated with both RFS (P = 0.008 and 0.0001) and DMFS (P = 0.019 and 0.001). CONCLUSIONS Gene expression profiling Class 2 result and SNB positivity were independently associated with recurrence and distant metastasis in primary CM patients. GEP testing may have additive prognostic utility in initial staging work-up of these patients.
Collapse
Affiliation(s)
- Jennifer Keller
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | | | | | - Ea‐sle Chang
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | - Ashaki D. Patel
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | - Maria Y. Hurley
- Department of DermatologySaint Louis UniversitySt. LouisMissouri
| | - Eddy C. Hsueh
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| |
Collapse
|
1400
|
O'Donnell JS, Hoefsmit EP, Smyth MJ, Blank CU, Teng MWL. The Promise of Neoadjuvant Immunotherapy and Surgery for Cancer Treatment. Clin Cancer Res 2019; 25:5743-5751. [PMID: 31040150 DOI: 10.1158/1078-0432.ccr-18-2641] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapies utilizing immune checkpoint inhibitors (ICI) have demonstrated durable efficacy in a proportion of patients with advanced/metastatic cancers. More recently, the use of ICIs for the adjuvant treatment of patients with surgically resectable melanoma has also demonstrated efficacy by improving relapse-free survival and in the case of ipilimumab (anti-CTLA-4) also improving overall survival. Although promising, the effective scheduling of surgery and immunotherapy and its duration is not well elucidated. Recent preclinical studies suggest that surgery followed by adjuvant therapy might be suboptimal as compared with an approach in which immunotherapy is applied before surgery (neoadjuvant immunotherapy). Encouraging findings from early-phase clinical trials in melanoma, non-small cell lung carcinoma, and glioblastoma support the idea that neoadjuvant immunotherapy might have improved clinical efficacy over an adjuvant application. In this review, we discuss the existing rationale for the use of neoadjuvant immunotherapy, its apparent strengths and weaknesses, and implications for the design of future clinical trials.
Collapse
Affiliation(s)
- Jake S O'Donnell
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Esmée P Hoefsmit
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mark J Smyth
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Christian U Blank
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Medical Oncology Department, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. .,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|