1
|
Veas Rodríguez J, Prieto A, Vilaprinyo E, Bonet M, Diez M, Salud A, Montal R. Surrogate endpoints in phase III randomized trials of advanced gastroesophageal cancer: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 201:104416. [PMID: 38871262 DOI: 10.1016/j.critrevonc.2024.104416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Overall survival (OS) is the most meaningful endpoint in clinical trials. However, owing to their limitations, surrogate endpoints are commonly used and validation studies are required to assess their reliability. Analysis of phase III randomized controlled trials (RCTs) of advanced gastroesophageal cancer (AGC) with > 100 patients, correlation coefficients (r), and determination coefficients (R²) between OS and surrogates were evaluated through meta-analyses. Progression-free survival (PFS), time to progression (TTP), and objective response rate (ORR) were examined to determine their correlations with OS. Analysis of 65 phase III RCTs (29,766 subjects) showed a moderate correlation between PFS/TTP and OS (r = 0.77, R² = 0.59), while ORR correlation was low (r = 0.56, R² = 0.31). Excluding immunotherapy trials improved the PFS/TTP and OS correlations (r = 0.83, R² = 0.70). These findings suggest the potential use of PFS/TTP in AGC phase III investigations, disregarding the use of ORR as a surrogate endpoint.
Collapse
Affiliation(s)
- Joel Veas Rodríguez
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Lleida, Spain; Department of Medical Oncology, Taunton and Somerset NHS Foundation Trust, Taunton, United Kingdom.
| | - Ana Prieto
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Ester Vilaprinyo
- Department of Basic Medical Sciences, University of Lleida, IRBLLEIDA, Lleida, Spain
| | - Marta Bonet
- Department of Radiation Oncology, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Marc Diez
- Department of Medical Oncology, Vall d' Hebron University Hospital, Barcelona, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Robert Montal
- Department of Medical Oncology, Arnau de Vilanova University Hospital, Lleida, Spain
| |
Collapse
|
2
|
Zhao YY, Ge HJ, Yang WT, Shao ZM, Hao S. Secretory breast carcinoma: clinicopathological features and prognosis of 52 patients. Breast Cancer Res Treat 2024; 203:543-551. [PMID: 37897648 DOI: 10.1007/s10549-023-07153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/29/2023] [Indexed: 10/30/2023]
Abstract
PURPOSE Secretory breast carcinoma is a rare histological subtype of invasive breast cancer and considered with an indolent clinical behavior. This study was conducted to analyze the clinicopathological features of patients with secretory breast carcinoma (SBC), explore the outcome, and compare the prognostic difference with invasive ductal breast carcinoma (IDC). METHODS AND MATERIALS: Patients with SBC diagnosed between 2006 and 2017 from Fudan University Shanghai Cancer Center were included in the study, excluding patients with previous malignant tumor history and incomplete clinical data or follow-up records. Peculiar clinicopathological and immunohistochemical features of the cases were fully described. Clinical data of 4979 cases of IDC were also evaluated during this period. After propensity score matching, prognostic analysis of SBCs and IDCs was calculated by Kaplan-Meier method and landmark analysis method. RESULTS The data of 52 patients diagnosed with SBC were identified from the pathological files. Among them, 47 patients were women, and 5 were men. The median age of the 52 SBCs was 46 years (mean, 48.1 years; range, 10-80 years). The tumor sizes ranged from 0.3 to 6.8 cm, with a mean of 3.5 cm. Eight patients (15.4%) had positive axillary lymph node involvement. The molecular classification was mostly triple-negative breast cancer (65.4%). Fluorescence in situ hybridization confirmed the presence of ETV6::NTRK3 rearrangement in 16 of 18 cases (88.9%). Furthermore, Kaplan-Meier survival analysis and landmark analysis demonstrated that there were no statistically significant differences in DFS and OS between SBC and IDC patients. CONCLUSION Although SBCs are generally associated with a favorable prognosis, our work exhibited that the clinicopathological features of SBC were partly different from former understandings, indicating that therapeutic procedure should be prudent. Further studies are necessary to fully identify the clinical behavior and predictive markers to improve diagnosis and management in this unique subtype of breast cancer.
Collapse
Affiliation(s)
- Yuan-Yuan Zhao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, People's Republic of China
| | - Hui-Juan Ge
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
| | - Wen-Tao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China.
- Institutes of Biomedical Science, Fudan University, Shanghai, People's Republic of China.
| | - Shuang Hao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
3
|
Xu C, Zhang S, Zhu GL, Yang KB, Zhang Y, Mao YP, Tang LL, Liu Q, Huang Y, Ma J. Disparities in positive results and dissemination of randomized controlled trials in immuno-oncology. Int Rev Immunol 2023; 42:91-100. [PMID: 35712868 DOI: 10.1080/08830185.2022.2088744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND This cross-sectional and longitudinal analysis aimed to demonstrate the disparities in positive results and dissemination patterns of randomized controlled trials (RCTs) in global immuno-oncology (IO). METHODS Phase II-IV RCTs with results reported by article publications registered on ClinicalTrials.gov in 2007-2018 studying immune checkpoint inhibitors (ICIs), adoptive cell transfer, cancer vaccines, and immune modulators were included. RESULTS Twenty-eight percent of trials were positive (72 of 258), most of which were pharma-sponsored and focused on ICI and multiple IO therapies in lung cancer, melanoma, and multiple cancer types. The recent period of trial start year, upfront registration, large sample size, high strictness score on corticosteroid/infection-related criteria, and survival endpoints were associated with positive results. Trials from Mainland China had a faster publication timeline of positive results but lacked study diversity or full reporting of negative results compared with US and multinational trials. Compared with phase II trials, phase III-IV trials had a higher average proportion of positive results (28.9% vs. 22.2%) and a more stable change over the past decade (23.65% vs. 49.24%). Positive trials yielded more secondary manuscripts (10 vs. 4), a shorter publication process of approximately two years (P < 0.001), and a superiority in the dissemination of journals with an h-index >90 (P < 0.001) compared with negative trials. CONCLUSION Disparities in positive result dissemination are widespread in IO RCTs and affected by trial features. We proposed improvements in upfront registration, procedural integrity, and adequate inclusion of rival trials reporting negative results within the earlier two years in future reviews.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shu Zhang
- Department of Radiation Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guang-Li Zhu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Kai-Bin Yang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Sun Yat-sen Global Health Institute, School of Public Health and Institute of State Governance, Sun Yat-sen University, Guangzhou, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qing Liu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ying Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
4
|
Vathiotis IA, Salichos L, Martinez-Morilla S, Gavrielatou N, Aung TN, Shafi S, Wong PF, Jessel S, Kluger HM, Syrigos KN, Warren S, Gerstein M, Rimm DL. Baseline gene expression profiling determines long-term benefit to programmed cell death protein 1 axis blockade. NPJ Precis Oncol 2022; 6:92. [PMID: 36522538 PMCID: PMC9755314 DOI: 10.1038/s41698-022-00330-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022] Open
Abstract
Treatment with immune checkpoint inhibitors has altered the course of malignant melanoma, with approximately half of the patients with advanced disease surviving for more than 5 years after diagnosis. Currently, there are no biomarker methods for predicting outcome from immunotherapy. Here, we obtained transcriptomic information from a total of 105 baseline tumor samples comprising two cohorts of patients with advanced melanoma treated with programmed cell death protein 1 (PD-1)-based immunotherapies. Gene expression profiles were correlated with progression-free survival (PFS) within consecutive clinical benefit intervals (i.e., 6, 12, 18, and 24 months). Elastic net binomial regression models with cross validation were utilized to compare the predictive value of distinct genes across time. Lasso regression was used to generate a signature predicting long-term benefit (LTB), defined as patients who remain alive and free of disease progression at 24 months post treatment initiation. We show that baseline gene expression profiles were consistently able to predict long-term immunotherapy outcomes with high accuracy. The predictive value of different genes fluctuated across consecutive clinical benefit intervals, with a distinct set of genes defining benefit at 24 months compared to earlier outcomes. A 12-gene signature was able to predict LTB following anti-PD-1 therapy with an area under the curve (AUC) equal to 0.92 and 0.74 in the training and validation set, respectively. Evaluation of LTB, via a unique signature may complement objective response classification and characterize the logistics of sustained antitumor immune responses.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| | - Leonidas Salichos
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Biological and Chemical Sciences, New York Institute of Technology, New York, USA
| | - Sandra Martinez-Morilla
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Niki Gavrielatou
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Thazin Nwe Aung
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Saba Shafi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Pok Fai Wong
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Shlomit Jessel
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Harriet M Kluger
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Konstantinos N Syrigos
- Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | | | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
Davar D, Zarour HM. Facts and Hopes for Gut Microbiota Interventions in Cancer Immunotherapy. Clin Cancer Res 2022; 28:4370-4384. [PMID: 35748749 PMCID: PMC9561605 DOI: 10.1158/1078-0432.ccr-21-1129] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/06/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023]
Abstract
Immune checkpoint inhibitors (ICI) targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) proteins transformed the management of advanced cancers. Many tumor-intrinsic factors modulate immunological and clinical responses to such therapies, but ample evidence also implicates the gut microbiome in responses. The gut microbiome, comprising the bacteria, archaea, fungi, and viruses that live in the human digestive tract, is an established determinant of host immunity, but its impact on response to ICI therapy in mice and humans with cancer has only recently been appreciated. Therapeutic interventions to optimize microbiota composition to improve immunotherapy outcomes show promise in mice and humans with cancer. In this review, we discuss the rationale for gut microbiome-based cancer therapies, the results from early-phase clinical trials, and possible future developments.
Collapse
Affiliation(s)
- Diwakar Davar
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hassane M. Zarour
- Department of Medicine and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
Avoidance of intramedullary violation in computer-assisted total knee arthroplasty lowers the incidence of periprosthetic joint infection compared with conventional total knee arthroplasty: A propensity score matching analysis of 5342 cases. Knee 2022; 35:164-174. [PMID: 35316778 DOI: 10.1016/j.knee.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2022] [Accepted: 03/03/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Conventional total knee arthroplasty (CONV-TKA) inevitably perturbs femoral medullary canal, disturbs medullary micro-architecture and increases blood loss and inflammatory responses. We hypothesized that avoidance of intramedullary violation may lower the incidence of periprosthetic joint infection (PJI). The aim of this study was to verify whether computer-assisted total knee arthroplasty (CAS-TKA) lowers the incidence of PJI as compared with CONV-TKA. METHODS A propensity score matching study of 5342 patients who underwent CAS-TKA (n = 1085) or CONV-TKA (n = 4257) for primary osteoarthritis of the knee from 2007 to 2015 in our institute was performed. Patients who underwent CAS-TKA were matched to those who received CONV-TKA at a 1:2 ratio according to demographics and comorbidities. PJI was defined according to the Musculoskeletal Infection Society diagnostic criteria from the 2013 International Consensus Meeting. RESULTS After controlling potential risk factors, the use of CAS-TKA resulted in a lower incidence of PJI as compared with CONV-TKA [adjusted hazard ratio (HR), 0.42; 95% confidence interval (CI), 0.18-0.99]. The same trend in PJI reduction was observed with the usage of CAS-TKA under sensitivity testing [HR, 0.33; 95% CI, 0.12-0.95]. The cumulative incidence of PJI was lower in the CAS-TKA group than the CONV-TKA group (log-rank test, p = 0.013). CONCLUSION Avoidance of intramedullary violation during TKA may play a pivotal role in lowering the incidence of PJI. The use of CAS-TKA can reduce the incidence of PJI, with a better survival rate in terms of being free of PJI, as compared with CONV-TKA.
Collapse
|
7
|
Ferrucci PF, Di Giacomo AM, Del Vecchio M, Atkinson V, Schmidt H, Schachter J, Queirolo P, Long GV, Stephens R, Svane IM, Lotem M, Abu-Amna M, Gasal E, Ghori R, Diede SJ, Croydon ES, Ribas A, Ascierto PA. KEYNOTE-022 part 3: a randomized, double-blind, phase 2 study of pembrolizumab, dabrafenib, and trametinib in BRAF-mutant melanoma. J Immunother Cancer 2021; 8:jitc-2020-001806. [PMID: 33361337 PMCID: PMC7768966 DOI: 10.1136/jitc-2020-001806] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background In the KEYNOTE-022 study, pembrolizumab with dabrafenib and trametinib (triplet) improved progression-free survival (PFS) versus placebo with dabrafenib and trametinib (doublet) without reaching statistical significance. Mature results on PFS, duration of response (DOR), and overall survival (OS) are reported. Methods The double-blind, phase 2 part of KEYNOTE-022 enrolled patients with previously untreated BRAFV600E/K-mutated advanced melanoma from 22 sites in seven countries. Patients were randomly assigned 1:1 to intravenous pembrolizumab (200 mg every 3 weeks) or placebo plus dabrafenib (150 mg orally two times per day) and trametinib (2 mg orally one time a day). Primary endpoint was PFS. Secondary endpoints were objective response rate, DOR, and OS. Efficacy was assessed in the intention-to-treat population, and safety was assessed in all patients who received at least one dose of study drug. This analysis was not specified in the protocol. Results Between November 30, 2015 and April 24, 2017, 120 patients were randomly assigned to triplet (n=60) or doublet (n=60) therapy. With 36.6 months of follow-up, median PFS was 16.9 months (95% CI 11.3 to 27.9) with triplet and 10.7 months (95% CI 7.2 to 16.8) with doublet (HR 0.53; 95% CI 0.34 to 0.83). With triplet and doublet, respectively, PFS at 24 months was 41.0% (95% CI 27.4% to 54.2%) and 16.3% (95% CI 8.1% to 27.1%); median DOR was 25.1 months (95% CI 14.1 to not reached) and 12.1 months (95% CI 6.0 to 15.7), respectively. Median OS was not reached with triplet and was 26.3 months with doublet (HR 0.64; 95% CI 0.38 to 1.06). With triplet and doublet, respectively, OS at 24 months was 63.0% (95% CI 49.4% to 73.9%) and 51.7% (95% CI 38.4% to 63.4%). Grade 3–5 treatment-related adverse events (TRAEs) occurred in 35 patients (58%, including one death) receiving triplet and 15 patients (25%) receiving doublet. Conclusion In BRAFV600E/K-mutant advanced melanoma, pembrolizumab plus dabrafenib and trametinib substantially improved PFS, DOR, and OS with a higher incidence of TRAEs. Interpretation of these results is limited by the post hoc nature of the analysis.
Collapse
Affiliation(s)
- Pier Francesco Ferrucci
- Cancer Biotherapy Unit, Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Anna Maria Di Giacomo
- Center for Immuno-Oncology, University Hospital of Siena; University of Siena, Siena, Italy
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Victoria Atkinson
- University of Queensland, and Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Woolloongabba, Queensland, Australia
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob Schachter
- Division of Oncology, Sheba Medical Centre, Tel HaShomer Hospital, Tel Aviv, Israel
| | - Paola Queirolo
- Divisione di Oncologia Medica del Melanoma, Sarcoma e Tumori Rari, European Institute of Oncology IRCCS, Milan, Italy
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore Hospital, Sydney, New South Wales, Australia.,Department of Medical Oncology and Translational Research, Mater Hospital, North Sydney, New South Wales, Australia
| | | | - Inge Marie Svane
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Michal Lotem
- Sharett Institute of Oncology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Eduard Gasal
- Global Drug Development, Oncology, Novartis, East Hanover, New Jersey, USA
| | - Razi Ghori
- Department of Clinical Oncology, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Scott J Diede
- Department of Clinical Oncology, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Elizabeth S Croydon
- Department of Clinical Oncology, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Antoni Ribas
- Department of Medicine, University of California Los Angeles and the Jonsson Comprehensive Cancer Center, Los Angeles, California, USA
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| | | |
Collapse
|
8
|
Leary A, Tan D, Ledermann J. Immune checkpoint inhibitors in ovarian cancer: where do we stand? Ther Adv Med Oncol 2021; 13:17588359211039899. [PMID: 34422119 PMCID: PMC8377306 DOI: 10.1177/17588359211039899] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
Numerous retrospective studies have demonstrated that the density of intra-tumoral immune cell infiltration is prognostic in epithelial ovarian cancer (OC). These observations together with reports of programmed death ligand-1 (PD-L1) expression in advanced OC provided the rationale for investigating the benefit of programmed death-1 (PD1) or PD-L1 inhibition in OC. Unfortunately clinical trials to date evaluating PD1/PD-L1 inhibition in patients with relapsed OC have been disappointing. In this review we will discuss early results from single agent PD1/PD-L1 inhibitors and the strategies to enhance benefit from immune-oncology agents in OC, including proposing anti-PD-L1 in combination with other agents (cytotoxics, anti-angiogenics, poly(ADP-ribose) polymerase. (PARP) inhibitors, targeted therapies or other immunotherapies), as well as evaluating these agents earlier in the disease course, or in biomarker selected patients.
Collapse
Affiliation(s)
- Alexandra Leary
- Institut Gustave Roussy, 114 rue Edouard Vaillant, Villejuif 94805, France, Université Paris-Saclay, INSERM U981, Villejuif, France
| | - David Tan
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Cancer Science Institute, National University of Singapore, Singapore
| | - Jonathan Ledermann
- UCL Cancer Institute, Cancer Research UK and UCL Trials Centre, London, UK
| |
Collapse
|
9
|
Fu J, Ding Z, Chen Q, Lin K, Liu H, Gao Y, Zeng Y, Li H, Shen F, Liu J. Distinct Impacts of Pre-Operative Antiviral Treatment on Post-Operative Outcomes of HBV-related Hepatocellular Carcinoma: A Landmark Analysis. J Cancer 2021; 12:170-180. [PMID: 33391413 PMCID: PMC7738838 DOI: 10.7150/jca.47125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The effect of anti-viral treatment (AVT) initiated before surgery (pre-operative AVT) on HBV-related hepatocellular carcinoma (HCC) has been controversial. This study aimed to elucidate the prognostic significance of pre-operative AVT for HCC patients who received hepatectomy. Materials and Methods: A large-scale retrospective study was conducted based on a cohort consisting of 1937 HBV-related HCC patients who underwent R0 liver resection between January 2011 and December 2012. Propensity score matching (PSM) method was adopted to balance covariates and landmark survival analyses were performed to visualize effects in different phases after surgery. Results: After PSM, a total of matched 744 patients (372 in each group) were recruited. The patients in the pre-operative AVT group had lower HBV-DNA loading levels and better recurrence-free survival (RFS) than those in the non-AVT group. The 1, 3, 5-year RFS rates of two groups were 67.3%, 49.0%, and 43.1% vs. 66.7%, 41.1% and 18.5%, respectively (P<0.001). Landmark survival analyses demonstrated that pre-operative AVT could improve RFS, and the effect was beginning to show after the first 12 months. There was no significant difference of overall survival (OS) between the two groups (P=0.543), and the landmark survival analyses indicated that pre-operative AVT could improve OS and this effect was beginning to show after 36 months. Additionally, multivariate Cox regression analyses revealed that larger tumor (>5cm), esophageal and gastric varices, lymph node metastasis were independent risk factors of RFS, and larger tumor (>5cm) and ascites were independent risk factors of OS. Conclusions: Pre-operative AVT could significantly improve the RFS, and could not improve short-term OS (< 36 months) but could better long-term survival of the patients with HBV-HCC after surgery.
Collapse
Affiliation(s)
- Jun Fu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Big Data Institute of Southeast Hepatobiliary Health Information, Fuzhou, China
| | - Zongren Ding
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Big Data Institute of Southeast Hepatobiliary Health Information, Fuzhou, China
| | - Qinjunjie Chen
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Kongying Lin
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Big Data Institute of Southeast Hepatobiliary Health Information, Fuzhou, China
| | - Hongzhi Liu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- The Big Data Institute of Southeast Hepatobiliary Health Information, Fuzhou, China
| | - Yuzhen Gao
- Department of Laboratory Medicine, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yongyi Zeng
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Haitao Li
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Feng Shen
- Department of Hepatic Surgery IV, the Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
10
|
van Zeijl MCT, de Wreede LC, van den Eertwegh AJM, Wouters MWJM, Jochems A, Schouwenburg MG, Aarts MJB, van Akkooi ACJ, van den Berkmortel FWPJ, de Groot JWB, Hospers GAP, Kapiteijn E, Piersma D, van Rijn RS, Suijkerbuijk KPM, Ten Tije AJ, van der Veldt AAM, Vreugdenhil G, van der Hoeven JJM, Haanen JBAG. Survival outcomes of patients with advanced melanoma from 2013 to 2017: Results of a nationwide population-based registry. Eur J Cancer 2020; 144:242-251. [PMID: 33373869 DOI: 10.1016/j.ejca.2020.11.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/15/2020] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND The treatment landscape has completely changed for advanced melanoma. We report survival outcomes and the differential impact of prognostic factors over time in daily clinical practice. METHODS From a Dutch nationwide population-based registry, patients with advanced melanoma diagnosed from 2013 to 2017 were analysed (n = 3616). Because the proportional hazards assumption was violated, a multivariable Cox model restricted to the first 6 months and a multivariable landmark Cox model from 6 to 48 months were used to assess overall survival (OS) of cases without missing values. The 2017 cohort was excluded from this analysis because of the short follow-up time. RESULTS Median OS of the 2013 and 2016 cohort was 11.7 months (95% confidence interval [CI]: 10.4-13.5) and 17.7 months (95% CI: 14.9-19.8), respectively. Compared with the 2013 cohort, the 2016 cohort had superior survival in the Cox model from 0 to 6 months (hazard ratio [HR] = 0.55 [95% CI: 0.43-0.72]) and in the Cox model from 6 to 48 months (HR = 0.68 [95% CI: 0.57-0.83]). Elevated lactate dehydrogenase levels, distant metastases in ≥3 organ sites, brain and liver metastasis and Eastern Cooperative Oncology Group performance score of ≥1 had stronger association with inferior survival from 0 to 6 months than from 6 to 48 months. BRAF-mutated melanoma had superior survival in the first 6 months (HR = 0.50 [95% CI: 0.42-0.59]). CONCLUSION(S) Prognosis for advanced melanoma in the Netherlands has improved from 2013 to 2016. Prognostic importance of most evaluated factors was higher in the first 6 months after diagnosis. BRAF-mutated melanoma was only associated with superior survival in the first 6 months.
Collapse
Affiliation(s)
- M C T van Zeijl
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, the Netherlands; Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, the Netherlands
| | - L C de Wreede
- Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, Leiden, 2333ZC, the Netherlands
| | - A J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, Location VU Medical Center (VUmc), Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
| | - M W J M Wouters
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, the Netherlands; Department of Surgical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, the Netherlands
| | - A Jochems
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, the Netherlands; Department of Medical Oncology, Haaglanden Medisch Centrum, Lijnbaan 32, Den Haag, 2512VA, the Netherlands
| | - M G Schouwenburg
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, Leiden, 2333AA, the Netherlands
| | - M J B Aarts
- Department of Medical Oncology, Maastricht University Medical Center, P. Debyelaan 25, Maastricht, 6229 HX, the Netherlands
| | - A C J van Akkooi
- Department of Surgical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, the Netherlands
| | - F W P J van den Berkmortel
- Department of Medical Oncology, Zuyderland Medical Center, Dr. H. van der Hoffplein 1, Sittard-Geleen, 6162BG, the Netherlands
| | - J W B de Groot
- Department of Medical Oncology, Isala Clinics, Dokter van Heesweg 2, Zwolle, 8025AB, the Netherlands
| | - G A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, Groningen, 9713GZ, the Netherlands
| | - E Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, the Netherlands
| | - D Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Koningsplein 1, Enschede, 7512KZ, the Netherlands
| | - R S van Rijn
- Department of Internal Medicine, Medical Center Leeuwarden, Henri Dunantweg 2, Leeuwarden, 8934AD, the Netherlands
| | - K P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Cancer Center, Heidelberglaan 100, Utrecht, 3584CX, the Netherlands
| | - A J Ten Tije
- Department of Internal Medicine, Amphia Hospital, Molengracht 21, Breda, 4818CK, the Netherlands
| | - A A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, Rotterdam, 3015CE, the Netherlands
| | - G Vreugdenhil
- Department of Internal Medicine, Maxima Medical Center, De Run 4600, Eindhoven, 5504DB, the Netherlands
| | - J J M van der Hoeven
- Department of Medical Oncology, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, 6525GA, the Netherlands
| | - J B A G Haanen
- Divisions of Medical Oncology and Molecular Oncology & Immunology, the Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066CX, the Netherlands.
| |
Collapse
|
11
|
Michielin O, Atkins MB, Koon HB, Dummer R, Ascierto PA. Evolving impact of long-term survival results on metastatic melanoma treatment. J Immunother Cancer 2020; 8:e000948. [PMID: 33037115 PMCID: PMC7549477 DOI: 10.1136/jitc-2020-000948] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Melanoma treatment has been revolutionized over the past decade. Long-term results with immuno-oncology (I-O) agents and targeted therapies are providing evidence of durable survival for a substantial number of patients. These results have prompted consideration of how best to define long-term benefit and cure. Now more than ever, oncologists should be aware of the long-term outcomes demonstrated with these newer agents and their relevance to treatment decision-making. As the first tumor type for which I-O agents were approved, melanoma has served as a model for other diseases. Accordingly, discussions regarding the value and impact of long-term survival data in patients with melanoma may be relevant in the future to other tumor types. Current findings indicate that, depending on the treatment, over 50% of patients with melanoma may gain durable survival benefit. The best survival outcomes are generally observed in patients with favorable prognostic factors, particularly normal baseline lactate dehydrogenase and/or a low volume of disease. Survival curves from melanoma clinical studies show a plateau at 3 to 4 years, suggesting that patients who are alive at the 3-year landmark (especially in cases in which treatment had been stopped) will likely experience prolonged cancer remission. Quality-of-life and mixture-cure modeling data, as well as metrics such as treatment-free survival, are helping to define the value of this long-term survival. In this review, we describe the current treatment landscape for melanoma and discuss the long-term survival data with immunotherapies and targeted therapies, discussing how to best evaluate the value of long-term survival. We propose that some patients might be considered functionally cured if they have responded to treatment and remained treatment-free for at least 2 years without disease progression. Finally, we consider that, while there have been major advances in the treatment of melanoma in the past decade, there remains a need to improve outcomes for the patients with melanoma who do not experience durable survival.
Collapse
Affiliation(s)
- Olivier Michielin
- Oncology Department, Precision Oncology Center, Lausanne, Switzerland
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Michael B Atkins
- Medical Oncology, Georgetown Lombardi Comprehensive Cancer Center and Oncology Academic Department, Georgetown University Medical Center, Washington, DC, USA
| | - Henry B Koon
- Clinical Research, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Paolo Antonio Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| |
Collapse
|
12
|
Quinn C, Garrison LP, Pownell AK, Atkins MB, de Pouvourville G, Harrington K, Ascierto PA, McEwan P, Wagner S, Borrill J, Wu E. Current challenges for assessing the long-term clinical benefit of cancer immunotherapy: a multi-stakeholder perspective. J Immunother Cancer 2020; 8:e000648. [PMID: 32661115 PMCID: PMC7359062 DOI: 10.1136/jitc-2020-000648] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 12/17/2022] Open
Abstract
Immuno-oncologics (IOs) differ from chemotherapies as they prime the patient's immune system to attack the tumor, rather than directly destroying cancer cells. The IO mechanism of action leads to durable responses and prolonged survival in some patients. However, providing robust evidence of the long-term benefits of IOs at health technology assessment (HTA) submission presents several challenges for manufacturers. The aim of this article was to identify, analyze, categorize, and further explore the key challenges that regulators, HTA agencies, and payers commonly encounter when assessing the long-term benefits of IO therapies. Insights were obtained from an international, multi-stakeholder steering committee (SC) and expert panels comprising of payers, economists, and clinicians. The selected individuals were tasked with developing a summary of challenges specific to IOs in demonstrating their long-term benefits at HTA submission. The SC and expert panels agreed that standard methods used to assess the long-term benefit of anticancer drugs may have limitations for IO therapies. Three key areas of challenges were identified: (1) lack of a disease model that fully captures the mechanism of action and subsequent patient responses; (2) estimation of longer-term outcomes, including a lack of agreement on ideal methods of survival analyses and extrapolation of survival curves; and (3) data limitations at the time of HTA submission, for which surrogate survival end points and real-world evidence could prove useful. A summary of the key challenges facing manufacturers when submitting evidence at HTA submission was developed, along with further recommendations for manufacturers in what evidence to produce. Despite almost a decade of use, there remain significant challenges around how best to demonstrate the long-term benefit of checkpoint inhibitor-based IOs to HTA agencies, clinicians, and payers. Manufacturers can potentially meet or mitigate these challenges with a focus on strengthening survival analysis methodology. Approaches to doing this include identifying reliable biomarkers, intermediate and surrogate end points, and the use of real-world data to inform and validate long-term survival projections. Wider education across all stakeholders-manufacturers, payers, and clinicians-in considering the long-term survival benefit with IOs is also important.
Collapse
Affiliation(s)
| | - Louis P Garrison
- CHOICE Institute, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | - Phil McEwan
- Centre for Health Economics, Swansea University, Swansea, UK
| | | | | | - Elise Wu
- Bristol-Myers Squibb, New York, New York, USA
| |
Collapse
|
13
|
Endpoint surrogacy in oncology Phase 3 randomised controlled trials. Br J Cancer 2020; 123:333-334. [PMID: 32451466 PMCID: PMC7403397 DOI: 10.1038/s41416-020-0896-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Endpoint surrogacy is an important concept in oncology trials. Using a surrogate endpoint like progression-free survival as the primary endpoint-instead of overall survival-would lead to a potential faster drug approval and therefore more cancer patients with an earlier opportunity to receive the newly approved drugs.
Collapse
|
14
|
Smoragiewicz M, Bogaerts J, Calvo E, Marabelle A, Perrone A, Seymour L, Shalabi A, Siu LL, Tabernero J, Giaccone G. Design and conduct of early clinical studies of immunotherapy agent combinations: recommendations from the task force on Methodology for the Development of Innovative Cancer Therapies. Ann Oncol 2019; 29:2175-2182. [PMID: 30202892 DOI: 10.1093/annonc/mdy398] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The Methodology for the Development of Innovative Cancer Therapies task force considered aspects of the design and conduct of early studies of combinations of immunotherapy agents during their 2018 meeting. The task force defined the relevant data to justify combination clinical trials, which includes a robust hypothesis for the combination, pre-clinical data with evidence of efficacy and an understanding of the pharmacodynamics effects of each agent, and ideally evidence of single agent activity. Evaluation of pharmacodynamic biomarkers is critical in early phase combination trials, and should be incorporated into trial objectives and go/no-go decisions. The task force also identified the need to develop assessment tools and end points that capture the unique patterns of tumour responses to immunotherapy, including pseudoprogression and hyperprogression. At least one additional tumour measurement before baseline and an early CT scan (at 4 weeks for example) would help define the incidence of hyperprogression, although a common definition is needed. Finally, the task force highlighted substantial redundancy and inefficiency in the combination immunotherapy space, and recommended the adoption of innovative trial designs.
Collapse
Affiliation(s)
- M Smoragiewicz
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada
| | | | - E Calvo
- START Madrid-Centro Integral Oncologico Clara Campal Hospital, Madrid, Spain
| | - A Marabelle
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, INSERM U1015, Villejuif, France
| | - A Perrone
- Translational Medicine, Merck & Co, Kenilworth
| | - L Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, Canada.
| | - A Shalabi
- The Anna-Maria Kellen Clinical Accelerator Cancer, Research Institute, New York, USA
| | - L L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Canada
| | - J Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - G Giaccone
- Georgetown University Medical Center, Washington, USA
| | | |
Collapse
|
15
|
Lauschke VM, Zhou Y, Ingelman-Sundberg M. Novel genetic and epigenetic factors of importance for inter-individual differences in drug disposition, response and toxicity. Pharmacol Ther 2019; 197:122-152. [PMID: 30677473 PMCID: PMC6527860 DOI: 10.1016/j.pharmthera.2019.01.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Individuals differ substantially in their response to pharmacological treatment. Personalized medicine aspires to embrace these inter-individual differences and customize therapy by taking a wealth of patient-specific data into account. Pharmacogenomic constitutes a cornerstone of personalized medicine that provides therapeutic guidance based on the genomic profile of a given patient. Pharmacogenomics already has applications in the clinics, particularly in oncology, whereas future development in this area is needed in order to establish pharmacogenomic biomarkers as useful clinical tools. In this review we present an updated overview of current and emerging pharmacogenomic biomarkers in different therapeutic areas and critically discuss their potential to transform clinical care. Furthermore, we discuss opportunities of technological, methodological and institutional advances to improve biomarker discovery. We also summarize recent progress in our understanding of epigenetic effects on drug disposition and response, including a discussion of the only few pharmacogenomic biomarkers implemented into routine care. We anticipate, in part due to exciting rapid developments in Next Generation Sequencing technologies, machine learning methods and national biobanks, that the field will make great advances in the upcoming years towards unlocking the full potential of genomic data.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
16
|
Ascierto PA, Marabelle A. How do immune checkpoint-targeted antibodies work? The need for improved pharmacokinetic evaluation in early phase studies. Ann Oncol 2018; 29:2157-2160. [PMID: 30307539 DOI: 10.1093/annonc/mdy420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- P A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italic.
| | - A Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif; INSERM U1015, Villejuif, France
| |
Collapse
|
17
|
Certainty within uncertainty: a qualitative study of the experience of metastatic melanoma patients undergoing pembrolizumab immunotherapy. Support Care Cancer 2018; 27:1845-1852. [DOI: 10.1007/s00520-018-4443-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/23/2018] [Indexed: 12/17/2022]
|
18
|
Ascierto PA, Dummer R. Immunological effects of BRAF+MEK inhibition. Oncoimmunology 2018; 7:e1468955. [PMID: 30228935 DOI: 10.1080/2162402x.2018.1468955] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/19/2022] Open
Abstract
Recent developments in immunotherapy have prolonged overall survival in metastatic melanoma with the possibility to reach a long-term benefit. Targeted therapies based on BRAF and MEK inhibition also seem to have a long-term beneficial effect, which is more evident in patients with favorable baseline characteristics, namely normal levels of lactate dehydrogenase, without brain metastases, and low tumor burden. This long-term benefit of targeted therapies might be related to an immune-modulation: indeed BRAF and MEK inhibitors affect tumor microenvironment and immune surveillance, and it has been shown that patients with complete response to targeted treatment have a pre-existing favorable immunologic signature.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland
| |
Collapse
|
19
|
Ferrara R, Pilotto S, Caccese M, Grizzi G, Sperduti I, Giannarelli D, Milella M, Besse B, Tortora G, Bria E. Do immune checkpoint inhibitors need new studies methodology? J Thorac Dis 2018; 10:S1564-S1580. [PMID: 29951307 PMCID: PMC5994495 DOI: 10.21037/jtd.2018.01.131] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 01/19/2018] [Indexed: 12/26/2022]
Abstract
Immune checkpoint inhibitors (ICI) have widely reshaped the treatment paradigm of advanced cancer patients. Although multiple studies are currently evaluating these drugs as monotherapies or in combination, the choice of the most accurate statistical methods, endpoints and clinical trial designs to estimate the benefit of ICI remains an unsolved methodological issue. Considering the unconventional patterns of response or progression [i.e., pseudoprogression, hyperprogression (HPD)] observed with ICI, the application in clinical trials of novel response assessment tools (i.e., iRECIST) able to capture delayed benefit of immunotherapies and/or to quantify tumor dynamics and kinetics over time is an unmet clinical need. In addition, the proportional hazard model and the conventional measures of survival [i.e., median overall or progression free survival (PFS) and hazard ratios (HR)] might usually result inadequate in the estimation of the long-term benefit observed with ICI. For this reason, innovative methodologies such as milestone analysis, restricted mean survival time (RMST), parametric models (i.e., Weibull distribution, weighted log rank test), should be systematically investigated in clinical trials in order to adequately quantify the fraction of patients who are "cured", represented by the tails of the survival curves. Regarding predictive biomarkers, in particular PD-L1 expression, the integration and harmonization of the existing assays are urgently needed to provide clinicians with reliable diagnostic tests and to improve patient selection for immunotherapy. Finally, developing original and high-quality study designs, such as adaptive or basket biomarker enriched clinical trials, included in large collaborative platforms with multiple active sites and cross-sector collaboration, represents the successful strategy to optimally assess the benefit of ICI in the next future.
Collapse
Affiliation(s)
- Roberto Ferrara
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Sara Pilotto
- U.O.C. Oncology, University of Verona, Comprehensive Cancer Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Mario Caccese
- U.O.C. Oncology, University of Verona, Comprehensive Cancer Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Giulia Grizzi
- U.O.C. Oncology, University of Verona, Comprehensive Cancer Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | | | | | - Benjamin Besse
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Giampaolo Tortora
- U.O.C. Oncology, University of Verona, Comprehensive Cancer Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Emilio Bria
- U.O.C. Oncology, University of Verona, Comprehensive Cancer Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
20
|
Katsoulakis E, Leeman JE, Lok BH, Shi W, Zhang Z, Tsai JC, McBride SM, Sherman EJ, Cohen M, Wong R, Ganly I, Lee NY, Riaz N. Long-term outcomes in oral cavity squamous cell carcinoma with adjuvant and salvage radiotherapy after surgery. Laryngoscope 2018; 128:2539-2545. [PMID: 29637571 DOI: 10.1002/lary.27191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/24/2018] [Accepted: 02/21/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Early-stage oral cavity squamous cell carcinoma (OCSCC) represents a heterogeneous group of patients, and locoregional recurrence rates are as high as 25% with surgery alone. Radiotherapy (RT) is typically reserved as part of salvage multimodality therapy after salvage surgery because it is generally thought that there is no significant detriment to salvage therapy. Our aim was to examine outcomes for recurrent OCSCC treated with salvage surgery and radiation and compare them to outcomes for patients treated with adjuvant postoperative RT upfront. METHODS We identified 425 patients with OCSCC treated with postoperative RT at our institution. The 5-year rates of local failure, locoregional failure (LRF), survival, and distant metastasis (DM) were the main outcome measures. We performed a landmark analysis and examined the same outcomes in the adjuvant versus salvage cohorts using Cox proportional hazards and Fine-Gray competing risk method. RESULTS The adjuvant cohort had higher tumor (T) (P < 0.0001) and nodal (N) (P < 0.0001) stage than the salvage cohort's stage at initial presentation. On multivariate analysis, a strategy of salvage RT experienced poorer overall survival (OS) compared to upfront adjuvant RT (hazard ratio [HR] 1.84; 95% confidence interval [CI], 1.26-2.70; P = 0.002). Moreover, salvage surgery followed by RT patients experienced increasing risk of LRF (HR 1.56; 95% CI, 1.18-2.06; P = 0.002) and (DM) (HR 1.53; 95% CI, 1.08-2.17; P = 0.02) on multivariate analysis. Additional analysis was performed excluding salvage cohort with advanced disease at initial presentation (T3-T4 and N2). Salvage RT treatment selection for early-stage OCSCC continued to experience significantly poorer OS as compared to adjuvant RT (HR 1.48; 95% CI, 1.002-2.19; P = 0.049). CONCLUSION Early-stage OCSCC patients who are observed and experienced recurrence requiring salvage therapy (surgery and RT) have worse oncologic outcomes than locally advanced patients receiving upfront adjuvant RT. Prospective randomized studies are needed to identify high-risk subset of early-stage OCSCC comparing adjuvant RT versus observation, followed by salvage surgery and RT at recurrence. LEVEL OF EVIDENCE 4. Laryngoscope, 2539-2545, 2018.
Collapse
Affiliation(s)
| | - Jonathan E Leeman
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Benjamin H Lok
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Weiji Shi
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Zhigang Zhang
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Jillian C Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Sean M McBride
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Eric J Sherman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Marc Cohen
- Department of Surgical Oncology ENT, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Richard Wong
- Department of Surgical Oncology ENT, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Ian Ganly
- Department of Surgical Oncology ENT, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, U.S.A
| |
Collapse
|
21
|
Ascierto PA, Daniele B, Hammers H, Hirsh V, Kim J, Licitra L, Nanda R, Pignata S. Perspectives in immunotherapy: meeting report from the "Immunotherapy Bridge", Napoli, November 30th 2016. J Transl Med 2017; 15:205. [PMID: 29020960 PMCID: PMC5637331 DOI: 10.1186/s12967-017-1309-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/30/2017] [Indexed: 12/26/2022] Open
Abstract
The complex interactions between the immune system and tumors lead the identification of key molecules that govern these interactions: immunotherapeutics were designed to overcome the mechanisms broken by tumors to evade immune destruction. After the substantial advances in melanoma, immunotherapy currently includes many other type of cancers, but the melanoma lesson is essential to progress in other type of cancers, since immunotherapy is potentially improving clinical outcome in various solid and haematologic malignancies. Monotherapy in pre-treated NSCLC is studied and the use of nivolumab, pembrolizumab and atezolizumab as second-line of advanced NSCLC is demonstrated as well as first line monotherapy and combination therapy in metastatic NSCLC studied. Patients with HNSCC have immunotherapeutic promises as well: the FDA recently approved moAbs targeting immune checkpoint receptors. Nivolumab in combination with ipilumumab showed acceptable safety and encouraging antitumor activity in metastatic renal carcinoma. HCCs have significant amounts of genomic heterogeneity and multiple oncogenic pathways can be activated: the best therapeutic targets identification is ongoing. The treatment of advanced/relapsed EOC remain clearly an unmet need: a better understanding of the relevant immuno-oncologic pathways and their corresponding biomarkers are required. UC is an immunotherapy-responsive disease: after atezolizumab, three other PD-L1/PD-L1 inhibitors (nivolumab, durvalumab, and avelumab) were approved for treatment of platinum-refractory metastatic urothelial carcinoma. Anti-PD-1/PD-L1 monotherapy is associated with a modest response rate in metastatic breast cancer; the addition of chemotherapy is associated with higher response rates. Immunotherapy safety profile is advantageous, although, in contrast to conventional chemotherapy: boosting the immune system leads to a unique constellation of inflammatory toxicities known as immune-related Adverse Events (irAEs) that may warrant the discontinuation of therapy and/or the administration of immunosuppressive agents. Research should explore better combination with less side effects, the right duration of treatments, combination or sequencing treatments with target therapies. At present, treatment decision is based on patient's characteristics.
Collapse
Affiliation(s)
- Paolo A. Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori “Fondazione G. Pascale”, Via Mariano Semmola, 80131 Naples, Italy
| | - Bruno Daniele
- Department of Oncology and Medical Oncology Unit, G. Rummo Hospital, Benevento, Italy
| | | | - Vera Hirsh
- McGill Department of Oncology, McGill University, Montreal, Canada
| | - Joseph Kim
- Medical Oncology, Yale School of Medicine, New Haven, CT USA
| | - Lisa Licitra
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Rita Nanda
- Section of Hematology–Oncology, Department of Medicine, The University of Chicago, Chicago, IL USA
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori “Fondazione G. Pascale”, Naples, Italy
| |
Collapse
|
22
|
Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, Seliger B, Marincola FM. Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer 2017. [PMID: 28623775 DOI: 10.1016/j.ejca.2017.01.035] [Citation(s) in RCA: 396] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer immunotherapy is now established as a powerful way to treat cancer. The recent clinical success of immune checkpoint blockade (antagonists of CTLA-4, PD-1 and PD-L1) highlights both the universal power of treating the immune system across tumour types and the unique features of cancer immunotherapy. Immune-related adverse events, atypical clinical response patterns, durable responses, and clear overall survival benefit distinguish cancer immunotherapy from cytotoxic cancer therapy. Combination immunotherapies that transform non-responders to responders are under rapid development. Current challenges facing the field include incorporating immunotherapy into adjuvant and neoadjuvant cancer therapy, refining dose, schedule and duration of treatment and developing novel surrogate endpoints that accurately capture overall survival benefit early in treatment. As the field rapidly evolves, we must prioritise the development of biomarkers to guide the use of immunotherapies in the most appropriate patients. Immunotherapy is already transforming cancer from a death sentence to a chronic disease for some patients. By making smart, evidence-based decisions in developing next generation immunotherapies, cancer should become an imminently treatable, curable and even preventable disease.
Collapse
Affiliation(s)
- Leisha A Emens
- Johns Hopkins University School of Medicine, Department of Oncology, Graduate Program in Pathobiology, Baltimore, MD 21287, USA.
| | - Paolo A Ascierto
- Istituto Nazionale Tumori Fondazione G. Pascale, Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Napoli, Italy
| | - Phillip K Darcy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, Australia
| | - Sandra Demaria
- Weill Cornell Medical College, Department of Radiation Oncology, New York, NY 10065, USA
| | - Alexander M M Eggermont
- Cancer Institute Gustave-Roussy, 114 Rue Edouard Vaillant, Villejuif/Paris-Sud 94800, France
| | - William L Redmond
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Barbara Seliger
- Martin Luther University, Institute for Medical Immunology, Magdeburger Str. 2, 06112 Halle, Germany
| | | |
Collapse
|
23
|
Abstract
Treatment options for patients with metastatic melanoma, and especially BRAF-mutant melanoma, have changed dramatically in the past 5 years, with the FDA approval of eight new therapeutic agents. During this period, the treatment paradigm for BRAF-mutant disease has evolved rapidly: the standard-of-care BRAF-targeted approach has shifted from single-agent BRAF inhibition to combination therapy with a BRAF and a MEK inhibitor. Concurrently, immunotherapy has transitioned from cytokine-based treatment to antibody-mediated blockade of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and, now, the programmed cell-death protein 1 (PD-1) immune checkpoints. These changes in the treatment landscape have dramatically improved patient outcomes, with the median overall survival of patients with advanced-stage melanoma increasing from approximately 9 months before 2011 to at least 2 years - and probably longer for those with BRAF-V600-mutant disease. Herein, we review the clinical trial data that established the standard-of-care treatment approaches for advanced-stage melanoma. Mechanisms of resistance and biomarkers of response to BRAF-targeted treatments and immunotherapies are discussed, and the contrasting clinical benefits and limitations of these therapies are explored. We summarize the state of the field and outline a rational approach to frontline-treatment selection for each individual patient with BRAF-mutant melanoma.
Collapse
|
24
|
Park HS, Sohn J, Kim SI, Park S, Park HS, Gho SG, Chung HC, Paik S, Kim GM. Effects of hormone receptor status on the durable response of trastuzumab-based therapy in metastatic breast cancer. Breast Cancer Res Treat 2017; 163:255-262. [PMID: 28243895 DOI: 10.1007/s10549-017-4175-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Trastuzumab-based treatment is the standard care for patients with HER2+ metastatic breast cancer (MBC). About 10% of HER2+ MBC showed a long-term durable response (progression-free survival, PFS > 3 years) to trastuzumab-based therapy. The aim of this study is to identify clinico-pathologic factors for a durable response to trastuzumab-based therapy in HER2-positive MBC. METHODS In the Yonsei Breast Cancer MBC Database, we identified 1218 MBC patients who were diagnosed from 2006 to 2015. Among them, 294 had HER2+ disease, and 153 received trastuzumab plus taxane chemotherapy as first-line treatment. Clinico-pathologic factors, such as hormone receptor (HR) status and metastatic sites, were reviewed. To evaluate a durable response, landmark analysis was performed. RESULTS The median follow-up time was 28 months (95% CI 4.4-83.0 months). Of 153 HER2+ patients, there were 73 HR- patients (47.7%), and bone was the most common metastatic site. The median PFS and overall survival (OS) were 12 and 39 months, respectively. HR- patients showed a tendency toward longer PFS (median, 13 vs. 11 months, P = 0.160) compared with HR+ patients. Patients with non-visceral metastases had longer median PFS and OS than those with visceral disease (median PFS, 15 vs. 11 months, P = 0.012; median OS, 75 vs. 34 months, P = 0.03). Landmark analysis at 9 months suggested that the PFS of HR- patients was significantly longer than that of HR+ patients (median, 19 vs. 9 months, P = 0.008). CONCLUSIONS Among patients with HER2+ MBC, HR status is a possible predictive biomarker of a durable response to trastuzumab-based therapy.
Collapse
Affiliation(s)
- Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Seung Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seho Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Seok Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seul Ghi Gho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Soonmyung Paik
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea.,Department of Biomedical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Gun Min Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea.
| |
Collapse
|
25
|
Simeone E, Grimaldi AM, Festino L, Giannarelli D, Vanella V, Palla M, Curvietto M, Esposito A, Palmieri G, Mozzillo N, Ascierto PA. Correlation between previous treatment with BRAF inhibitors and clinical response to pembrolizumab in patients with advanced melanoma. Oncoimmunology 2017; 6:e1283462. [PMID: 28405510 PMCID: PMC5384373 DOI: 10.1080/2162402x.2017.1283462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022] Open
Abstract
The optimal sequencing of targeted treatment and immunotherapy in the treatment of advanced melanoma is a key question and prospective studies to address this are ongoing. Previous observations suggest that treating first with targeted therapy may select for more aggressive disease, meaning that patients may not gain full benefit from subsequent immunotherapy. In a single-center retrospective analysis, we investigated whether response to pembrolizumab was affected by previous BRAF inhibitor therapy. A total of 42 patients with metastatic cutaneous or mucosal melanoma who had received previous treatment with ipilimumab were treated with pembrolizumab as part of the Italian expanded access program. Sixteen of these patients had BRAF-mutated melanoma and had also been previously treated with a BRAF inhibitor (vemurafenib or dabrafenib), while 26 had BRAF wild-type melanoma (no previous BRAF inhibitor). Patients with BRAF-mutant melanoma who were previously treated with BRAF inhibitors had a significantly lower median progression-free survival (3 [2.3-3.7] versus not reached [2-8+] mo; p = 0.001) and disease control rate (18.6% versus 65.4%; p = 0.005) than patients with BRAF wild-type, while there was also a trend toward a lower response rate (assessed using immune-related response criteria) although this was not significantly different between groups (12.5% versus 36.4%; p = 0.16). These data are consistent with previous reports that BRAF inhibitor therapy may affect subsequent response to immunotherapy.
Collapse
Affiliation(s)
- Ester Simeone
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Antonio Maria Grimaldi
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Lucia Festino
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | | | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Marco Palla
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Marcello Curvietto
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Assunta Esposito
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| | - Giuseppe Palmieri
- Unit of Melanoma and Sarcoma Surgery, Istituto Nazionale Tumori Fondazione, Napoli, Italy
| | - Nicola Mozzillo
- Unit of Melanoma and Sarcoma Surgery, Istituto Nazionale Tumori Fondazione, Napoli, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U., Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| |
Collapse
|