151
|
Wong CK, Lam TH, Liao SY, Lau YM, Tse HF, So BYF. Immunopathogenesis of Immune Checkpoint Inhibitor Induced Myocarditis: Insights from Experimental Models and Treatment Implications. Biomedicines 2023; 11:biomedicines11010107. [PMID: 36672615 PMCID: PMC9855410 DOI: 10.3390/biomedicines11010107] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Despite the extraordinary success of immune checkpoint inhibitors (ICIs) in cancer treatment, their use is associated with a high incidence of immune-related adverse events (IRAEs), resulting from therapy-related autoimmunity against various target organs. ICI-induced myocarditis is one of the most severe forms of IRAE, which is associated with risk of hemodynamic compromise and mortality. Despite increasing recognition and prompt treatment by clinicians, there remain significant gaps in knowledge regarding the pathophysiology, diagnosis and treatment of ICI-induced myocarditis. As the newly emerged disease entity is relatively rare, it is challenging for researchers to perform studies involving patients at scale. Alternatively, mouse models have been developed to facilitate research understanding of the pathogenesis of ICI-induced myocarditis and drug discovery. Transgenic mice with immune checkpoint genes knocked out allow induction of myocarditis in a highly reproducible manner. On the other hand, it has not been possible to induce ICI-induced myocarditis in wild type mice by injecting ICIs monotherapy alone. Additional interventions such as combinational ICI, tumor inoculation, cardiac sarcomere immunization, or cardiac irradiation are necessary to mimic the underlying pathophysiology in human cancer patients and to induce ICI-induced myocarditis successfully. This review focuses on the immunopathogenesis of ICI-induced myocarditis, drawing insights from human studies and animal models, and discusses the potential implications for treatment.
Collapse
Affiliation(s)
- Chun-Ka Wong
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tsun-Ho Lam
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Song-Yan Liao
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yee-Man Lau
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
| | - Benjamin Y. F. So
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Correspondence: ; Tel.: +852-2255-3111
| |
Collapse
|
152
|
Pedersen S, Holmstroem RB, von Heymann A, Tolstrup LK, Madsen K, Petersen MA, Haslund CA, Ruhlmann CH, Schmidt H, Johansen C, Svane IM, Ellebaek E. Quality of life and mental health in real-world patients with resected stage III/IV melanoma receiving adjuvant immunotherapy. Acta Oncol 2023; 62:62-69. [PMID: 36645166 DOI: 10.1080/0284186x.2023.2165449] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Treatment with immune checkpoint inhibitors (ICI) has expanded into the adjuvant setting enhancing the importance of knowledge on the immune-related toxicities and their impact on health-related quality of life (HRQoL). Large phase 3 trials of patients with resected Stage III/IV melanoma found no effect on HRQoL during adjuvant immunotherapy. This study investigates how HRQoL was affected during and after adjuvant immunotherapy in a real-world setting. METHODS Patients with resected melanoma treated with adjuvant nivolumab from 2018 to 2021 in Denmark were identified using the Danish Metastatic Melanoma Database (DAMMED). The study was performed as a nationwide cross-sectional analysis as a questionnaire consisting of six different validated questionnaires on HRQoL, cognitive function, fatigue, depression, fear of recurrence, and decision regret was sent to all patients in March 2021. To evaluate HRQoL during and after adjuvant treatment, patients were divided into groups depending on their treatment status when answering the questionnaire; patients in active treatment for 0-6 months, patients in active treatment for >6 months, patients who ended treatment 0-6 months ago, and patients who ended treatment >6 months ago. RESULTS A total of 271/412 (66%) patients completed the questionnaire. Patients who ended therapy 0-6 months ago had the lowest HRQoL and had more fatigue. Patients in active treatment for >6 months had lower HRQoL and more fatigue than patients who started treatment 0-6 months ago. Patients ending therapy >6 months ago had higher HRQoL and less fatigue compared to patients who ended therapy 0-6 months ago. Multivariable analysis showed an association between HRQoL and treatment status, comorbidity, civil status, and employment status. CONCLUSIONS Adjuvant nivolumab may affect some aspects of QoL, but the influence seems temporary. Patient characteristics, such as civil status, employment status, and comorbidity were associated with HRQoL.
Collapse
Affiliation(s)
- Sidsel Pedersen
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
| | - Rikke B Holmstroem
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
| | - Annika von Heymann
- Department of Oncology, Cancer Survivorship and Late Effects Research Center (CASTLE), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Kasper Madsen
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
| | - Morten Aagaard Petersen
- Department of Geriatrics and Palliative Medicine GP, Palliative Care Research Unit, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | | - Christina H Ruhlmann
- Department of Oncology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Christoffer Johansen
- Department of Oncology, Cancer Survivorship and Late Effects Research Center (CASTLE), Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Inge Marie Svane
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
| | - Eva Ellebaek
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Copenhagen University Hospital, Herlev and Gentofte, Copenhagen, Denmark
| |
Collapse
|
153
|
Shen X, Huang S, Xiao H, Zeng S, Liu J, Ran Z, Xiong B. Efficacy and safety of PD-1/PD-L1 plus CTLA-4 antibodies ± other therapies in lung cancer: a systematic review and meta-analysis. Eur J Hosp Pharm 2023; 30:3-8. [PMID: 34497128 PMCID: PMC9811552 DOI: 10.1136/ejhpharm-2021-002803] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/24/2021] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To investigate the efficacy and safety of programmed cell death 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) plus cytotoxic T lymphocyte antigen-4 (CTLA-4) antibodies ± other therapies in patients with advanced lung cancer. METHODS In accordance with the retrieval strategy, we searched electronic databases for randomised controlled trials testing PD-1/PD-L1 plus CTLA-4 antibodies in patients with lung cancer; RR (for objective response rate (ORR), overall survival (OS), progression-free survival (PFS), and immune-related adverse events (irAEs)) from individual studies were calculated and pooled by using random-effects models or fixed-effects models; heterogeneity and publication bias analyses were also performed, using Review Manager 5.3 and Stata 15.1 for statistical analysis. RESULTS We included six studies. Four different immune checkpoint inhibitors (nivolumab, pembrolizumab, durvalumab, tremelimumab) were used. Dual checkpoint inhibitors ± other therapies for advanced lung cancer showed significant improvements in ORR (RR 1.49, 95% CI 1.11 to 1.98; p=0.007), OS (HR 0.72, 95% CI 0.63 to 0.83; p<0.00001), and PFS (HR 0.72, 95% CI 0.63 to 0.82; p<0.00001). The subgroup analyses were consistent with the pooled results. The PD-L1 ≥1% (HR 0.67, 95% CI 0.54 to 0.82; p<0.0001) subgroup differences indicated a statistically significant subgroup effect, but the PD-L1 <1% subgroup (HR 0.88, 95% CI 0.75 to 1.05; p=0.15) was not statistically significant. The incidence of adverse events (grade ≥3) was lower than that of the control group (RR 0.90, 95% CI 0.80 to 1.02; p=0.09), but was not significant. CONCLUSIONS PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitors ± other therapies can improve the ORR, OS and PFS of patients with advanced or metastatic lung cancer, but the incidence of adverse reactions is high although generally tolerable. PROSPERO REGISTRATION CRD42020149216.
Collapse
Affiliation(s)
- Xiang Shen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shangke Huang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Hua Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shan Zeng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiexing Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhuolan Ran
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Xiong
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
154
|
Singh G, Thakur N, Kumar U. RAS: Circuitry and therapeutic targeting. Cell Signal 2023; 101:110505. [PMID: 36341985 DOI: 10.1016/j.cellsig.2022.110505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/05/2022] [Accepted: 10/21/2022] [Indexed: 11/26/2022]
Abstract
Cancer has affected the lives of millions worldwide and is truly regarded as a devastating disease process. Despite advanced understanding of the genomic underpinning of cancer development and progression, therapeutic challenges are still persistent. Among all the human cancers, around 33% are attributed to mutations in RAS oncogene, a crucial component of the signaling pathways. With time, our understanding of RAS circuitry has improved and now the fact that it activates several downstream effectors, depending on the type and grades of cancer has been established. The circuitry is controlled via post-transcriptional mechanisms and frequent distortions in these mechanisms lead to important metabolic as well as immunological states that favor cancer cells' growth, survival, plasticity and metastasis. Therefore, understanding RAS circuitry can help researchers/clinicians to develop novel and potent therapeutics that, in turn, can save the lives of patients suffering from RAS-mutant cancers. There are many challenges presented by resistance and the potential strategies with a particular focus on novel combinations for overcoming these, that could move beyond transitory responses in the direction of treatment. Here in this review, we will look at how understanding the circuitry of RAS can be put to use in making strategies for developing therapeutics against RAS- driven malignancies.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, NH-05, Ludhiana - Chandigarh State Hwy, Sahibzada Ajit Singh Nagar, Punjab 140413, India
| | - Neelam Thakur
- Department of Biosciences (UIBT), Chandigarh University, NH-05, Ludhiana - Chandigarh State Hwy, Sahibzada Ajit Singh Nagar, Punjab 140413, India; Department of Zoology, Sardar Patel University, Vallabh Government College Campus, Paddal, Kartarpur, Mandi, Himachal Pradesh 175001, India.
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH09, Ghaziabad, Uttar Pradesh 201015, India.
| |
Collapse
|
155
|
Jacques JP, Valadares LP, Moura AC, Oliveira MRF, Naves LA. Frequency and clinical characteristics of hypophysitis and hypopituitarism in patients undergoing immunotherapy - A systematic review. Front Endocrinol (Lausanne) 2023; 14:1091185. [PMID: 36875457 PMCID: PMC9974646 DOI: 10.3389/fendo.2023.1091185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE To describe the frequency of hypophysitis and hypopituitarism in cancer patients who are undergoing antineoplastic treatment with immunotherapy, as well as to describe the clinical, epidemiological, and demographic characteristics of these patients. METHODS A systematic search of the literature in PubMed, Embase, Web of Science, ClinicalTrials.gov and Cochrane Controlled Register of Trials took place on May 8 and 9, 2020. Randomized and nonrandomized clinical trials, cohort studies, case-control studies, case series and case reports were included. RESULTS A total of 239 articles were obtained, in which 963 cases of hypophysitis and 128 cases of hypopituitarism were found in a treated population of 30,014 individuals (3.20% and 0.42% of the evaluated population, respectively). In the cohort studies, the incidence of hypophysitis and hypopituitarism ranged from 0% to 27.59% and from 0% to 17.86%, respectively. In the non-randomized clinical trials, the incidence of hypophysitis and hypopituitarism ranged from 0% to 25% and from 0% to 14.67%, and in randomized clinical trials from 0% to 16.2% and from 0% to 33.33%. The most common hormonal changes were in the corticotrophic, thyrotrophic and gonadotrophic axes. The main magnetic resonance imaging (MRI) findings were enlargement of the pituitary gland and enhanced contrast uptake. The main symptoms presented by patients with hypophysitis were fatigue and headache. CONCLUSION The present review reported a frequency of hypophysitis and hypopituitarism of 3.20% and 0.42%, respectively, in the evaluated population. The clinical-epidemiological characteristics of patients affected by hypophysitis were also described. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/, identifier CRD42020175864.
Collapse
Affiliation(s)
- Juliana Prudêncio Jacques
- Faculty of Health Science, University of Brasilia, Brasilia, Brazil
- *Correspondence: Juliana Prudêncio Jacques,
| | - Luciana Pinto Valadares
- Department of Internal Medicine and Endocrinology, Sarah Network of Rehabilitation Hospitals, Brasília, Brazil
| | | | | | | |
Collapse
|
156
|
Cohen AO, Woo SH, Zhang J, Cho J, Ruiz ME, Gong J, Du R, Yarygina O, Jafri DZ, Bachelor MA, Finlayson MO, Soni RK, Hayden MS, Owens DM. Tbc1d10c is a selective, constitutive suppressor of the CD8 T-cell anti-tumor response. Oncoimmunology 2022; 11:2141011. [PMID: 36338148 PMCID: PMC9635554 DOI: 10.1080/2162402x.2022.2141011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cancer immunotherapy approaches target signaling pathways that are highly synonymous between CD4 and CD8 T-cell subsets and, therefore, often stimulate nonspecific lymphocyte activation, resulting in cytotoxicity to otherwise healthy tissue. The goal of our study was to identify intrinsic modulators of basic T lymphocyte activation pathways that could discriminately bolster CD8 anti-tumor effector responses. Using a Tbc1d10c null mouse, we observed marked resistance to a range of tumor types conferred by Tbc1d10c deficiency. Moreover, tumor-bearing Tbc1d10c null mice receiving PD-1 or CTLA-4 monotherapy exhibited a 33% or 90% cure rate, respectively. While Tbc1d10c was not expressed in solid tumor cells, Tbc1d10c disruption selectively augmented CD8 T-cell activation and cytotoxic effector responses and adoptive transfer of CD8 T cells alone was sufficient to recapitulate Tbc1d10c null tumor resistance. Mechanistically, Tbc1d10c suppressed CD8 T-cell activation and anti-tumor function by intersecting canonical NF-κB pathway activation via regulation of Map3k3-mediated IKKβ phosphorylation. Strikingly, none of these cellular or molecular perturbations in the NF-κB pathway were featured in Tbc1d10c null CD4 T cells. Our findings identify a Tbc1d10c-Map3k3-NF-κB signaling axis as a viable therapeutic target to promote CD8 T-cell anti-tumor immunity while circumventing CD4 T cell-associated cytotoxicity and NF-κB activation in tumor cells.
Collapse
Affiliation(s)
- Adrienne O. Cohen
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA
| | - Seung-Hyun Woo
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Discovery Biology Division, Velia Therapeutics, San Diego, CA, USA
| | - Junya Zhang
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA
| | - Jiyoon Cho
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Global Safety Assurance, Reckitt Benckiser Inc., Montvale, NJ, USA
| | - Marlon E. Ruiz
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Olink Proteomics, Los Angeles, CA90045, USA
| | - Jianli Gong
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Processing Cell Sciences, Merck & Co., Inc, Kenilworth, NJ, USA
| | - Rong Du
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA
| | - Olga Yarygina
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA
| | - Danya Z. Jafri
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA
| | - Michael A. Bachelor
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Boston Scientific, Center for Biological Innovation, Global Preclinical Sciences, Marlborough, MA, USA
| | - Michael O. Finlayson
- Department of Systems Biology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY, USA,Simons Foundation, New York, NY10010, USA
| | - Rajesh K. Soni
- Proteomics & Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY10032, USA
| | - Matthew S. Hayden
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - David M. Owens
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY10032, USA,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, NY, USA,CONTACT David M. Owens Russ Berrie Medical Science Pavilion, 1150 St. Nicholas Ave., Room 312A, New York, NY10032
| |
Collapse
|
157
|
Functional Gene Expression Signatures from On-Treatment Tumor Specimens Predict Anti-PD1 Blockade Response in Metastatic Melanoma. Biomolecules 2022; 13:biom13010058. [PMID: 36671443 PMCID: PMC9855743 DOI: 10.3390/biom13010058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Functional gene expression signatures (FGES) from pretreatment biopsy samples have been used to predict the responses of metastatic melanoma to immune checkpoint blockade (ICB) therapies. However, there are no predictive FGE signatures from patients receiving treatment. Here, using the Elastic Net Regression (ENLR) algorithm, we analyzed transcriptomic and matching clinical data from a dataset of patients with metastatic melanoma treated with ICB therapies and produced an FGE signature for pretreatment (FGES-PRE) and on-treatment (FGES-ON). Both the FGES-PRE and FGES-ON signatures are validated in three independent datasets of metastatic melanoma as the validation set, achieving area under the curve (AUC) values of 0.44-0.81 and 0.82-0.83, respectively. Then, we combined all test samples and obtained AUCs of 0.71 and 0.82 for the FGES-PRE and FGES-ON signatures, respectively. The FGES-ON signatures had a higher predictive value for prognosis than the FGES-PRE signatures. The FGES-PRE and FGES-ON signatures were divided into high- and low-risk scores using the signature score mean value. Patients with a high FGE signature score had better survival outcomes than those with low scores. Overall, we determined that the FGES-ON signature is an effective biomarker for metastatic melanoma patients receiving ICB therapy. This work would provide an important theoretical basis for applying FGE signatures derived from on-treatment tumor samples to predict patients' therapeutic response to ICB therapies.
Collapse
|
158
|
Sobrero AF, Pastorino A, Zalcberg JR. You're Cured Till You're Not: Should Disease-Free Survival Be Used as a Regulatory or Clinical End Point for Adjuvant Therapy of Cancer? J Clin Oncol 2022; 40:4044-4047. [PMID: 36315927 DOI: 10.1200/jco.22.01531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Alberto F Sobrero
- IRCCS Ospedale Policlinico San Martino, Medical Oncology Unit 1, Genova, Italy
| | | | - John R Zalcberg
- School of Public Health, Monash University and Department of Medical Oncology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
159
|
Li AA, Li F, Lan M, Zhang Y, Xie D, Yan MY. A novel endoplasmic reticulum stress-related lncRNA prognostic risk model for cutaneous melanoma. J Cancer Res Clin Oncol 2022; 148:3227-3241. [PMID: 35687183 DOI: 10.1007/s00432-022-04086-y] [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: 04/06/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Endoplasmic reticulum stress (ERS) and long non-coding RNAs (lncRNAs) are important in melanoma development and progression. This study aimed to explore the prognostic value of ERS-associated lncRNA profiles in cutaneous melanoma (CM). METHODS The Cancer Genome Atlas (TCGA) provides the raw data of CM. GSEA website was used to obtain ERS-related genes, and mRNA and LncRNA co-expression network were used to obtain ERS-related lncRNAs. A Lasso regression analysis was used to identify a prognostic risk model for the composition of ERS-related lncRNAs. Patients were divided into high- and low-risk groups based on the model's risk score. The researchers then compared the two groups' survival rates, immune infiltration, chemotherapeutic drug sensitivity, and immune checkpoint gene expression. RESULTS Thirty-nine ERS-related lncRNAs were discovered to be prognostic. A prognostic risk model made up of ten ERS-related lncRNAs was discovered. Patients in the low-risk group had a better prognosis than those in the high-risk group. An examination of tumor microenvironment revealed that risk scores correlated with immune cell infiltration in eight cases. Dacarbazine, paclitaxel, and cisplatin, three chemotherapy drugs, were more sensitive in the low-risk group than in the high-risk group. CONCLUSION This study identified a risk model of ten ERS-related lncRNAs that have significant prognostic value in CM and could help guide clinical treatment.
Collapse
Affiliation(s)
- An-An Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Fan Li
- Ji'an College, Ji'an, Jiangxi, People's Republic of China
| | - Min Lan
- Department of Orthopedic Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Yu Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Dong Xie
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Mei-Ying Yan
- Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Donghu District, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
160
|
Malaty MM, Amarasekera AT, Li C, Scherrer-Crosbie M, Tan TC. Incidence of immune checkpoint inhibitor mediated cardiovascular toxicity: A systematic review and meta-analysis. Eur J Clin Invest 2022; 52:e13831. [PMID: 35788986 DOI: 10.1111/eci.13831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) are a novel class of anti-cancer therapy becoming increasingly associated with fatal cardiovascular toxicities (CVTs). The aim is to determine the incidence of CVTs in cohorts treated with ICIs as sole anti-cancer therapy. METHODS A systematic literature search of scientific and medical databases was performed using PRISMA principles to identify relevant cohorts (PROSPERO registration CRD42021272470). Data for specific CVTs (pericardial disease, myocarditis, heart failure, arrhythmia, myocardial infarction/ischaemia and angina), CVT-related death and CV risk factors were extracted. Presence of CVTs in ICI-monotherapy versus combination-ICI therapy, and programmed death 1/programmed death ligand 1- (PD1/PDL1-) versus cytotoxic T-lymphocyte-associated protein 4- (CTLA4-) inhibitor groups were dichotomised and meta-analysed using random-effect models. RESULTS Forty-eight studies (11,207 patients) were identified, from which 146 CVTs were observed (incidence 1.30%). ICI-monotherapy led to more CVTs than combination therapy (119/9009; 1.32% vs. 18/2086; 0.86%). Across monotherapies, PD1/PDL1-inhibitors had lower incidence of CVTs compared to CTLA4-inhibitors (62/6950; 0.89% vs. 57/2059; 2.77%). Based on eight studies that were meta-analysed, no significant difference was observed comparing monotherapy versus combination-ICI therapy (RR-0.69, 95% CI -1.47 to 0.09) for all CVTs, or PD1/PDL1- to CTLA4-inhibitors (RR-0.27, 95% CI -2.06 to 1.53), for all CVTs including CVT-death. CV risk factors could not be attributed to an ICI group as data was population based rather than individual based. CONCLUSION ICI-mediated CVTs are rare and potentially fatal. The role of CV risk factors in their development remains unclear.
Collapse
Affiliation(s)
- Michael M Malaty
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Anjalee Thanuja Amarasekera
- School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia.,Westmead Applied Research Centre (WARC), Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Cindy Li
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy C Tan
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia.,School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia
| |
Collapse
|
161
|
Anestopoulos I, Kyriakou S, Tragkola V, Paraskevaidis I, Tzika E, Mitsiogianni M, Deligiorgi MV, Petrakis G, Trafalis DT, Botaitis S, Giatromanolaki A, Koukourakis MI, Franco R, Pappa A, Panayiotidis MI. Targeting the epigenome in malignant melanoma: Facts, challenges and therapeutic promises. Pharmacol Ther 2022; 240:108301. [PMID: 36283453 DOI: 10.1016/j.pharmthera.2022.108301] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
Malignant melanoma is the most lethal type of skin cancer with high rates of mortality. Although current treatment options provide a short-clinical benefit, acquired-drug resistance highlights the low 5-year survival rate among patients with advanced stage of the disease. In parallel, the involvement of an aberrant epigenetic landscape, (e.g., alterations in DNA methylation patterns, histone modifications marks and expression of non-coding RNAs), in addition to the genetic background, has been also associated with the onset and progression of melanoma. In this review article, we report on current therapeutic options in melanoma treatment with a focus on distinct epigenetic alterations and how their reversal, by specific drug compounds, can restore a normal phenotype. In particular, we concentrate on how single and/or combinatorial therapeutic approaches have utilized epigenetic drug compounds in being effective against malignant melanoma. Finally, the role of deregulated epigenetic mechanisms in promoting drug resistance to targeted therapies and immune checkpoint inhibitors is presented leading to the development of newly synthesized and/or improved drug compounds capable of targeting the epigenome of malignant melanoma.
Collapse
Affiliation(s)
- I Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - S Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - V Tragkola
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - I Paraskevaidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - E Tzika
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | | | - M V Deligiorgi
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - G Petrakis
- Saint George Hospital, Chania, Crete, Greece
| | - D T Trafalis
- Laboratory of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - S Botaitis
- Department of Surgery, Alexandroupolis University Hospital, Democritus University of Thrace School of Medicine, Alexandroupolis, Greece
| | - A Giatromanolaki
- Department of Pathology, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - M I Koukourakis
- Radiotherapy / Oncology, Radiobiology & Radiopathology Unit, Department of Medicine, School of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - R Franco
- Redox Biology Centre, University of Nebraska-Lincoln, Lincoln, NE, USA; School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - A Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - M I Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
| |
Collapse
|
162
|
Yin L, Liu X, Wu J, Yang J, Wang J, Dou H, Hou Y. LS-007 inhibits melanoma growth via inducing apoptosis and cell cycle arrest and regulating macrophage polarization. Melanoma Res 2022; 32:419-427. [PMID: 36094494 DOI: 10.1097/cmr.0000000000000853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
LS-007, an inhibitor of cyclin-dependent kinase 9 (CDK9), exhibits potential antitumor activity against chronic lymphocytic leukemia and ovarian cancer, but its effect on melanoma and tumor microenvironment (TME) has not been reported yet. This study aimed to investigate the role of LS-007 in B16F10 melanoma and relevant mechanisms. LS-007 significantly inhibited viability and induced apoptosis of B16F10 cells in a dose-dependent manner, which were accompanied with the increased ratio of Bax to Bcl-2 and decreased Mcl-1 mRNA level. Western blot analysis showed that LS-007 increased the expression of cleaved caspase-3 and poly ADP-ribose polymerase (PARP). Furthermore, flow cytometry analysis and qRT-PCR results showed that LS-007 treatment resulted in cell cycle arrest by changing cell cycle-related gene expression. Notably, in vivo evaluation showed that LS-007 significantly decreased the weight and volume of tumor and the expression of Ki67, promoted the expression of iNOS and inhibited the expression of CD206, suggesting that LS-007 might inhibit tumor growth by suppressing polarization of macrophages into tumor-associated macrophages (TAMs) in the TME. The increase in M1/M2 treated with LS-007 detected by flow cytometry hinted that macrophages were polarized towards an antitumor phenotype. In addition, LS-007 induced higher apoptotic rate of B16F10 cells when co-cultured B16F10 with BMDMs. LS-007 has inhibitory effects on B16F10 cells in vivo and in vitro via inducing apoptosis, cell cycle arrest, and changing macrophage function in the TME.
Collapse
Affiliation(s)
- Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jinjin Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jingjing Yang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Jiali Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
163
|
Keller J, Stern S, Chang SC, Marcus R, Weiss J, Nassoiy S, Christopher W, Fischer T, Essner R. Predicting Regional Lymph Node Recurrence in the Modern Age of Tumor-Positive Sentinel Node Melanoma: The Role of the First Postoperative Ultrasound. Ann Surg Oncol 2022; 29:8469-8477. [PMID: 35989390 DOI: 10.1245/s10434-022-12345-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The Multicenter Selective Lymphadenectomy Trial II (MSLT-II) led to a change in the management of tumor-positive sentinel lymph nodes (SLNs) from completion node dissection (CLND) to nodal observation. This study aimed to evaluate prognostic factors for predicting sentinel node basin recurrence (SNBR) using data from MSLT-II trial participants. METHODS In MSLT-II, 1076 patients were treated with observation. Patients were included in the current study if they had undergone a post-sentinel node basin ultrasound (PSNB-US) within 4 months after surgery. The study excluded patients with positive SLN by reverse transcription-polymerase chain reaction (RT-PCR) or incomplete SLN pathologic data. Primary tumor, patient, PSNB-US, and SLN characteristics were evaluated. Multivariable regression analyses were performed to determine independent prognostic factors associated with SNBR. RESULTS The study enrolled 737 patients: 193 (26.2%) patients with SNBR and 73 (9.9%) patients with first abnormal US. The patients with an abnormal first US were more likely to experience SNBR (23.8 vs. 5.0%). In the multivariable analyses, increased risk of SNBR was associated with male gender (adjusted hazard ratio [aHR], 1.38; 95% confidence interval [CI], 1.00-1.9; p = 0.049), increasing Breslow thickness (aHR, 1.10; 95% CI, 1.01-1.2; p = 0.038), presence of ulceration (aHR, 1.93; 95% CI, 1.42-2.6; p < 0.001), sentinel node tumor burden greater than 1 mm (aHR, 1.91; 95% CI, 1.10-3.3; p = 0.022), lymphovascular invasion (aHR, 1.53; 95% CI, 1.00-2.3; p = 0.048), and presence of abnormal PSNB-US (aHR, 4.29; 95% CI, 3.02-6.1; p < 0.001). CONCLUSIONS The first postoperative US together with clinical and pathologic factors may play an important role in predicting SNBR.
Collapse
Affiliation(s)
- Jennifer Keller
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Stacey Stern
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | | | - Rebecca Marcus
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Jessica Weiss
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Sean Nassoiy
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | | | - Trevan Fischer
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA
| | - Richard Essner
- Providence Saint John's Cancer Institute, Santa Monica, CA, USA.
| |
Collapse
|
164
|
Hooiveld-Noeken JS, Eggen AC, Rácz E, de Vries EG, Reyners AK, Jalving M. Towards less mutilating treatments in patients with advanced non-melanoma skin cancers by earlier use of immune checkpoint inhibitors. Crit Rev Oncol Hematol 2022; 180:103855. [DOI: 10.1016/j.critrevonc.2022.103855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022] Open
|
165
|
Xie R, Wu J, Shang B, Bi X, Cao C, Guan Y, Shi H, Shou J. Deaths and adverse events from adjuvant therapy with immune checkpoint inhibitors in solid malignant tumors: A systematic review and network meta-analysis. CANCER INNOVATION 2022; 1:293-304. [PMID: 38089086 PMCID: PMC10686117 DOI: 10.1002/cai2.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/13/2022] [Accepted: 09/28/2022] [Indexed: 10/15/2024]
Abstract
Background By prolonging overall survival and reducing disease recurrence rates, immune checkpoint inhibitors (ICIs) are an emerging adjuvant therapy option for patients with resectable malignant tumors. However, the safety profile (deaths and adverse events [AEs]) of adjuvant ICIs has not been fully described. Methods We searched the literature for phase III randomized clinical trials that compared PD-1, PD-L1, and CTLA-4 inhibitors in solid malignant tumors. Incidences of death, discontinuation, AEs of any cause, treatment-related adverse events (TRAEs), and immune-related adverse events (IRAEs) were extracted for the network meta-analysis. Network meta-analyses with low incidence and poor convergence are reported as incidences with 95% confidence intervals (95% CIs). Results Ten randomized clinical trials that included 9243 patients who received ICI adjuvant therapy were eligible. In total, 21 deaths due to TRAEs were recorded, with an overall incidence of 0.40% (95% CI: 0.26-0.61). The treatment-related mortality rates for ipilimumab (0.76%, 95% CI: 0.31-1.55) and atezolizumab (0.56%, 95% CI: 0.18-1.31) were higher than for pembrolizumab (0.24%, 95% CI: 0.10-0.56) and nivolumab (0.30%, 95% CI: 0.08-0.77). The most frequent causes of death were associated with the gastrointestinal (0.10%, 95% CI: 0.04-0.24) and pulmonary (0.08%, 95% CI: 0.03-0.21) systems. Compared with the control arm, we found that nivolumab (odds ratio [OR]: 2.73, 95% CI: 0.49-15.85) and atezolizumab (OR: 12.43, 95% CI: 2.42-78.48) caused the fewest grade ≥3 TRAEs and IRAEs. Commonly reported IRAEs of special interest were analyzed, and two agents were found to have IRAEs with incidences >10%, i.e., hepatitis for atezolizumab (14.80%, 95% CI: 12.53-17.32) and hypophysitis for ipilimumab (13.53%, 95% CI: 11.38-15.90). Conclusions Ipilimumab and atezolizumab were correlated with higher treatment-related death rates than pembrolizumab and nivolumab, in which the gastrointestinal and pulmonary systems were mostly involved. Regarding severe TRAEs and IRAEs, nivolumab and atezolizumab are likely to be the safest agent, respectively. This study will guide clinical practice for ICI adjuvant therapies.
Collapse
Affiliation(s)
- Ruiyang Xie
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jie Wu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Bingqing Shang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xingang Bi
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chuanzhen Cao
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Youyan Guan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hongzhe Shi
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianzhong Shou
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
166
|
Colucci M, D’Alonzo V, Santangelo F, Miracco C, Valente M, Maio M, Di Giacomo AM. Successful Targeting of CTLA-4 in a Melanoma Clinical Case: A Long-Term "One Stop Therapeutic Shop". Onco Targets Ther 2022; 15:1409-1415. [PMID: 36457762 PMCID: PMC9707535 DOI: 10.2147/ott.s367389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/12/2022] [Indexed: 02/04/2024] Open
Abstract
The anti-Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) monoclonal antibody ipilimumab was the first in-class immune-checkpoint inhibitor (ICI) approved for the treatment of melanoma patients. Initially approved for metastatic cutaneous melanoma, treatment with ipilimumab subsequently demonstrated to significantly improve recurrence free survival (RFS) in fully resected, high-risk, stage III melanoma patients. Therapeutic use of ipilimumab has also allowed the initial identification and characterization of unconventional clinical and radiological patterns of response (ie, tumor flare, pseudo-progression) that may occur during ICI therapy, unlike chemotherapy or targeted therapy. As a result, the standard Response Evaluation Criteria In Solid Tumors (RECIST) and the World Health Organization (WHO) criteria conventionally utilized to assess responses to chemo/targeted therapy have been initially replaced by the immune-related (ir) Response Criteria (irRC) and then by the irRECIST, that encompass all patterns of response typical of ICI therapy, being key for the optimal comprehensive management of treated patients. Here, we report a paradigmatic clinical case of a long-term survival in a stage III melanoma patient, experiencing tumor flares during adjuvant treatment with ipilimumab, and an untreated disease relapse several years after ending therapy.
Collapse
Affiliation(s)
| | | | | | - Clelia Miracco
- Department of Pathology, University of Siena, Siena, Italy
| | - Monica Valente
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital, Siena, Italy
| | - Michele Maio
- University of Siena, Siena, Italy
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital, Siena, Italy
- NIBIT Foundation Onlus, Genoa, Italy
| | - Anna Maria Di Giacomo
- University of Siena, Siena, Italy
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital, Siena, Italy
- NIBIT Foundation Onlus, Genoa, Italy
| |
Collapse
|
167
|
Kiuru M, Li Q, Zhu G, Terrell JR, Beroukhim K, Maverakis E, Keegan THM. Melanoma in women of childbearing age and in pregnancy in California, 1994-2015: a population-based cohort study. J Eur Acad Dermatol Venereol 2022; 36:2025-2035. [PMID: 35870141 PMCID: PMC9560982 DOI: 10.1111/jdv.18458] [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: 10/07/2021] [Accepted: 07/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Melanoma is one of the most common malignancies during pregnancy. There is debate regarding the impact of pregnancy on the prognosis of melanoma. Recent large population-based studies from the United States are lacking. OBJECTIVES To determine the characteristics and survival of women with pregnancy-associated melanoma. METHODS This population-based, retrospective cohort study used California Cancer Registry data linked with state-wide hospitalization and ambulatory surgery data to identify 15-44-year-old female patients diagnosed with melanoma in 1994-2015, including pregnant patients. Multivariable logistic regression compared demographic and clinical characteristics between pregnant and non-pregnant women with melanoma. Multivariable cox proportional hazards regression models assessed melanoma-specific and overall survival. RESULTS We identified 13 108 patients, of which 1406 were pregnant. Pregnancy-associated melanoma was more frequent in Hispanic compared to non-Hispanic White women. Melanoma occurring post-partum was associated with greater tumour thickness (2.01-4.00 vs. 0.01-1.00 mm, odds ratio 1.75, 95% confidence interval: 1.03-2.98). There were otherwise no significant differences between pregnant and non-pregnant women. Worse survival was associated with Asian, Black and Native American race/ethnicity (vs. non-Hispanic White), lower neighbourhood socio-economic status, public insurance, tumour site, greater tumour thickness and lymph node involvement, but not pregnancy. CONCLUSIONS Melanoma occurring post-partum was associated with greater tumour thickness, but pregnancy status did not affect survival after melanoma. Race/ethnicity, socio-economic status and health insurance impacted survival, emphasizing the importance of reducing health disparities.
Collapse
Affiliation(s)
- M Kiuru
- Department of Dermatology, University of California Davis, Sacramento, California, USA
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, California, USA
| | - Q Li
- Center for Oncology Hematology Outcomes Research and Training (COHORT) and Division of Hematology and Oncology, University of California Davis, Sacramento, California, USA
| | - G Zhu
- Department of Dermatology, University of California Davis, Sacramento, California, USA
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - J R Terrell
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | - K Beroukhim
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | - E Maverakis
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | - T H M Keegan
- Center for Oncology Hematology Outcomes Research and Training (COHORT) and Division of Hematology and Oncology, University of California Davis, Sacramento, California, USA
| |
Collapse
|
168
|
Eggermont AMM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, Dalle S, Haydon A, Meshcheryakov A, Khattak A, Carlino MS, Sandhu S, Larkin J, Puig S, Ascierto PA, Rutkowski P, Schadendorf D, Boers-Sonderen M, Di Giacomo AM, van den Eertwegh AJM, Grob JJ, Gutzmer R, Jamal R, van Akkooi ACJ, Lorigan P, Grebennik D, Krepler C, Marreaud S, Suciu S, Robert C. Five-Year Analysis of Adjuvant Pembrolizumab or Placebo in Stage III Melanoma. NEJM EVIDENCE 2022; 1:EVIDoa2200214. [PMID: 38319852 DOI: 10.1056/evidoa2200214] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
BACKGROUND: In the previously reported primary analyses of this phase 3 trial, 12 months of adjuvant pembrolizumab resulted in significantly longer recurrence- and distant metastasis-free survival than placebo in patients with resected high-risk stage III melanoma. To confirm the stability of these benefits, longer-term data were needed. METHODS: We randomly assigned 1019 patients to receive 200 mg of pembrolizumab or placebo intravenously every 3 weeks for a total of 18 doses (approximately 1 year) and had previously reported data with a 15-, 36-, and 42-month median follow-up. We now report data at a median follow-up of 4.9 years. We report a number of outcomes, including recurrence-free survival in the overall population and in the subgroup of patients with cancer who were positive for the programmed death-ligand 1 (PD-L1). Distant metastasis-free survival was a secondary end point. RESULTS: In the overall intention-to-treat population, pembrolizumab was still associated with longer recurrence-free survival than placebo (5-year rate of recurrence-free survival, 55.4% [95% confidence interval (CI), 50.8 to 59.8] vs. 38.3% [95% CI, 33.9 to 42.7]; hazard ratio for recurrence or death, 0.61 [95% CI, 0.51 to 0.72]) and a longer distant metastasis-free survival (5-year rate of distant metastasis-free survival, 60.6% [95% CI, 56.0 to 64.9] vs. 44.5% [95% CI, 39.9 to 48.9]; hazard ratio for distant metastasis or death, 0.62 [95% CI, 0.52 to 0.75]). Similar findings were obtained in the subgroup of 853 patients with PD-L1–positive tumors. CONCLUSIONS: The 5-year analysis of adjuvant therapy with pembrolizumab resulted in a sustained improvement in the long-term recurrence- and distant metastasis-free survival compared with placebo in patients with resected stage III melanoma. (Funded by Merck & Co., Inc.; ClinicalTrials.gov number, NCT02362594, and EudraCT number, 2014-004944-37.)
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Comprehensive Cancer Center Munich, München, Germany
- Princess Máxima Center and University Medical Center Utrecht, Netherlands
| | - Michal Kicinski
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Christian U Blank
- Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Mario Mandala
- University of Perugia, Santa Maria della Misericordia Hospital, Italy
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, Mater and Royal North Shore Hospitals, Sydney, VIC, Australia
| | - Victoria Atkinson
- Princess Alexandra Hospital, University of Queensland, Brisbane, QLD, Australia
| | - Stéphane Dalle
- Lyon Civic Hospital Cancer Institute, Cancer Research Centre of Lyon, Lyon University, France
| | | | - Andrey Meshcheryakov
- Federal State Budgetary Institution "Russian Oncology Scientific Centre named after N.N. Blokhin RAMS," Moscow, Russian Federation
| | - Adnan Khattak
- Fiona Stanley Hospital/University of Western Australia, Perth, WA, Australia
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia, and University of Sydney, Sydney, VIC, Australia
| | | | | | - Susana Puig
- Hospital Clínic de Barcelona, University of Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale," Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Dirk Schadendorf
- University Hospital Essen, Essen, Germany
- German Cancer Consortium, Heidelberg, Germany
| | | | - Anna Maria Di Giacomo
- Center for Immuno-Oncology and Medical Oncology and Immunotherapy Unit, University Hospital of Siena, Italy
| | | | - Jean-Jacques Grob
- Aix-Marseille University, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, France
| | - Ralf Gutzmer
- Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Germany
| | - Rahima Jamal
- Centre Hospitalier de l'Université de Montréal, Centre de Recherche du CHUM, Quebec, QC, Canada
| | | | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | - Sandrine Marreaud
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Stefan Suciu
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Caroline Robert
- Gustave Roussy Cancer Campus Grand Paris and University Paris-Saclay, Villejuif, France
| |
Collapse
|
169
|
Khattak MA, Luke JJ, Long GV, Ascierto PA, Rutkowski P, Schadendorf D, Robert C, Grob JJ, de la Cruz Merino L, Del Vecchio M, Spagnolo F, Mackiewicz J, Chiarion-Sileni V, Carlino MS, Mohr P, De Galitiis F, Ross MI, Eroglu Z, Chen K, Jiang R, Fukunaga-Kalabis M, Krepler C, Eggermont AMM, Kirkwood JM. Adjuvant pembrolizumab versus placebo in resected high-risk stage II melanoma: Health-related quality of life from the randomized phase 3 KEYNOTE-716 study. Eur J Cancer 2022; 176:207-217. [PMID: 36202690 DOI: 10.1016/j.ejca.2022.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/08/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Adjuvant pembrolizumab significantly improved recurrence-free survival (RFS) versus placebo in resected stage IIB and IIC melanoma in the phase 3 KEYNOTE-716 study. Health-related quality of life (HRQoL) results are reported. METHODS Patients were randomly assigned 1:1 to pembrolizumab 200 mg (2 mg/kg, patients ≥12 to <18 years) Q3W or placebo for ≤17 cycles or until disease recurrence, unacceptable toxicity, or withdrawal. Change from baseline in EORTC QLQ-C30 global health status (GHS)/quality of life (QoL) was a prespecified exploratory end point. Change in EORTC QLQ-C30 functioning, symptom, and single-item scales, and EQ-5D-5L visual analog scale (VAS) were also summarized. Primary analyses were performed at week 48 to ensure adequate completion/compliance. The HRQoL population comprised patients who received ≥1 dose of treatment and completed ≥1 assessment. RESULTS The HRQoL population included 969 patients (pembrolizumab, n = 483; placebo, n = 486). Compliance at week 48 was ≥80% for both instruments. EORTC QLQ-C30 GHS/QoL, physical functioning, role functioning, and EQ-5D-5L VAS scores were stable from baseline to week 48 in both arms, with no clinically meaningful decline observed. Scores did not differ significantly between pembrolizumab and placebo. EORTC QLQ-C30 GHS/QoL, physical functioning, role functioning, and EQ-5D-5L VAS scores remained stable through week 96 in both arms. CONCLUSIONS HRQoL was stable with adjuvant pembrolizumab, with no clinically meaningful decline observed. Change from baseline in HRQoL was similar between arms. These results, in conjunction with the improved RFS and manageable safety previously reported, support the use of adjuvant pembrolizumab for high-risk stage II melanoma.
Collapse
Affiliation(s)
- Muhammad A Khattak
- Fiona Stanley Hospital and Edith Cowan University, Perth, WA, Australia.
| | - Jason J Luke
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale," Naples, Italy
| | - Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Dirk Schadendorf
- Comprehensive Cancer Center, Universitaetsklinikum Essen and German Cancer Consortium, Essen, Germany
| | - Caroline Robert
- Dermatology Committee, Gustave Roussy, Université Paris-Sud, Villejuif, France
| | | | | | | | | | | | | | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Westmead and Blacktown Hospitals, Sydney, NSW, Australia
| | - Peter Mohr
- Elbe Kliniken Buxtehude, Buxtehude, Germany
| | | | - Merrick I Ross
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zeynep Eroglu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ke Chen
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | - Alexander M M Eggermont
- University Medical Center Utrecht and Princess Maxima Center, Utrecht, Netherlands; Comprehensive Cancer Center Munich, Munich, Germany
| | - John M Kirkwood
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
170
|
Kobeissi I, Tarhini AA. Systemic adjuvant therapy for high-risk cutaneous melanoma. Ther Adv Med Oncol 2022; 14:17588359221134087. [PMID: 36324735 PMCID: PMC9619267 DOI: 10.1177/17588359221134087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Cutaneous melanoma continues to increase in incidence and poses a significant mortality risk. Surgical excision of melanoma in its early stages is often curative. However, patients with resected stages IIB-IV are considered at high risk for relapse and death from melanoma where systemic adjuvant therapy is indicated. The long-studied high-dose interferon-α was shown to improve relapse-free survival (RFS) and overall survival (OS) but is no longer in use. Adjuvant therapy with ipilimumab at 10 mg/kg (ipi10) demonstrated significant RFS and OS improvements but at a high cost in terms of toxicity, while adjuvant ipilimumab 3 mg/kg was shown to be equally effective and less toxic. More recently, the adjuvant therapy for resected stages III-IV melanoma in clinical practice has changed in favor of nivolumab, pembrolizumab, and BRAF-MEK inhibitors dabrafenib plus trametinib (for BRAF mutant melanoma) based on significant improvements in RFS as compared to ipi10 (nivolumab and pembrolizumab) and placebo (dabrafenib plus trametinib). For resected stages IIB-IIC melanoma, pembrolizumab achieved regulatory approval in the United States based on significant RFS benefits. In this article, we review completed and ongoing phase III adjuvant therapy trials. We also briefly discuss neoadjuvant therapy for locoregionally advanced melanoma. Finally, we explore recent studies on predictive and prognostic melanoma biomarkers in the adjuvant setting.
Collapse
Affiliation(s)
- Iyad Kobeissi
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | |
Collapse
|
171
|
Kakish HH, Ahmed FA, Elshami M, Loftus AW, Hoehn RS, Ammori JB, Ocuin LM, Winter JM, Bordeaux JS, Mangla A, Rothermel LD. Trends in Melanoma Phase 3 Clinical Trials since 2010: Is there Hope for Advanced Melanoma Therapies beyond Approved Treatment Mechanisms? Cancers (Basel) 2022; 14:5184. [PMID: 36358601 PMCID: PMC9658976 DOI: 10.3390/cancers14215184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Several drugs and treatment modalities are under investigation to improve current melanoma therapy options. This review profiles the trends in clinical trial investment in late-stage melanoma, and anticipates what changes are expected in melanoma treatment, with a focus on exploratory drug mechanisms. METHODS We reviewed nine international clinical trial databases for registered, interventional, and phase 3 cutaneous melanoma clinical trials since 2010. RESULTS 73 trials studied drug therapies in late-stage (stage III and IV) melanoma. Exploratory mechanisms were investigated in 32% (23/73) of the late-stage melanoma drug therapy trials. Most exploratory drug trials include immunotherapy drug mechanisms (15/23 trials). Two exploratory mechanisms showed promise: the anti-LAG3 antibody, relatlimab, and the hapten modified vaccine, MVax. Many (52%) trials of exploratory mechanisms are ongoing including the use of adoptive cell transfer immunotherapies, dendritic cell vaccine therapy, and histone deacetylase (HDAC) inhibitors, among others. CONCLUSIONS Since most clinical trials focus on previously approved drug mechanisms, it is likely that paradigm-changing treatments will involve these therapies being used in new treatment contexts or combinations. Only 2 exploratory drug mechanisms studied since 2010 have achieved promising results in the phase 3 setting, though many other trials are ongoing at this time.
Collapse
Affiliation(s)
- Hanna H. Kakish
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Fasih Ali Ahmed
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Mohamedraed Elshami
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Alexander W. Loftus
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Richard S. Hoehn
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - John B. Ammori
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Lee M. Ocuin
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jordan M. Winter
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Jeremy S. Bordeaux
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Ankit Mangla
- Division of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Luke D. Rothermel
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
172
|
Long-Term Toxicities of Immune Checkpoint Inhibitor (ICI) in Melanoma Patients. Curr Oncol 2022; 29:7953-7963. [PMID: 36290906 PMCID: PMC9600354 DOI: 10.3390/curroncol29100629] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
ICI therapy has greatly improved patient outcomes in melanoma, but at the cost of immune-related adverse events (irAEs). Data on the chronicity of irAEs, especially in real-world settings, are currently limited. We performed a retrospective chart review of 161 adult patients with melanoma treated with at least one cycle of ICI regimen in the adjuvant or metastatic setting: 129 patients received PD-1 inhibitor monotherapy and 32 received dual immunotherapy. Patients were grouped by duration of irAE: permanent (no complete resolution), long-term (resolution over a period ≥ 6 months), transient (resolution over a period < 6 months), or no irAEs. A total of 283 irAEs were reported in the whole patient population. Sixty-six (41.0%) patients developed permanent irAEs, fifteen (9.3%) experienced long-term irAEs as their longest-lasting toxicity, thirty-four (21.1%) developed transient irAEs only, and forty-six (28.6%) experienced no irAEs. Permanent irAEs occurred in 21 (65.6%) patients treated with dual immunotherapy and in 45 (34.9%) patients treated with monotherapy. The majority of permanent irAEs were endocrine-related (36.0%) or skin-related (32.4%). Grade 3-4 permanent irAEs occurred in 20 (12.4%) patients and included toxicities such as adrenal insufficiency, myocarditis, and myelitis. Fifty-three (32.9%) patients were still requiring treatment for long-term or permanent irAEs 6 months or more following the completion of ICI therapy, including twenty-four patients on thyroid hormone replacement and twenty-two on oral steroids. ICI treatment was temporarily interrupted for 64 (22.6%) irAEs and permanently discontinued due to irAEs in 38 patients (13.6% of irAEs, 23.6% of patients); additionally, 4 (2.5%) patients died of irAEs. Our findings show that ICI treatment in melanoma is associated with a wide range of toxicities that can be permanent and may have long-lasting impacts on patients, which should therefore be discussed when obtaining consent for treatment.
Collapse
|
173
|
Rhodin KE, Fimbres DP, Burner DN, Hollander S, O’Connor MH, Beasley GM. Melanoma lymph node metastases - moving beyond quantity in clinical trial design and contemporary practice. Front Oncol 2022; 12:1021057. [PMID: 36411863 PMCID: PMC9675405 DOI: 10.3389/fonc.2022.1021057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/29/2022] [Indexed: 09/10/2023] Open
Abstract
The presence of lymph node metastases is a well-studied prognostic factor for cutaneous melanoma. Characterization of melanoma lymph node metastases and their association with survival in multiple, large observational studies has led to recognition of the following high-risk features: quantity of lymph node metastases (number of nodes), size of the nodal tumor deposit (in mm), and extracapsular extension. Despite increasing utilization of these features in the design of randomized clinical trials, in addition to their role in contemporary clinical decision-making, current staging systems lag behind, only accounting for the quantity of lymph nodes with metastases. Herein, we review the prognostic role of melanoma lymph node metastases and their high-risk features, current reporting standards, how such features have been utilized in practice-changing trials, and best practices for future clinical trial design and clinical decision-making.
Collapse
Affiliation(s)
- Kristen E. Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | | | - Shayna Hollander
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Margaret H. O’Connor
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Georgia M. Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
174
|
Meevassana J, Anothaisatapon K, Subbalekha S, Kamolratanakul S, Siritientong T, Ruangritchankul K, Pungrasami P, Hamill KJ, Angsapatt A, Kitkumthorn N. BRAF V600E Immunohistochemistry Predicts Prognosis of Patients with Cutaneous Melanoma in Thai population. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2022; 10:e4605. [PMID: 36299811 PMCID: PMC9592364 DOI: 10.1097/gox.0000000000004605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
UNLABELLED The BRAF V600E mutation in the Thai population has been identified in a considerable percentage of people with cutaneous melanoma. The objectives of this study were to determine the prevalence of this mutation in cutaneous melanomas, conduct a clinicopathological association analysis with the BRAF V600E mutation, and develop a treatment strategy for patients with this mutation that would take advantage of the medications currently available to treat them. METHODS Anti-BRAF V600E (clone VE1) immunohistochemistry was performed on 50 pathological samples of cutaneous melanoma after excluding the samples with a low amount of pathologic tissue, a lack of clinical data' and poor follow-up. BRAF V600E expression DNA sequencing was performed to confirm the results of several cases. RESULTS Anti-BRAF V600E antibody positivity was noted in 56% (28/50) of cutaneous melanoma cases. DNA sequencing results were consistent with immunohistochemistry results. In cutaneous melanoma, the BRAF V600E mutation was significantly associated with adverse prognosis of patients, including reduced overall survival and disease-free survival. CONCLUSIONS An increased prevalence of the BRAF V600E mutation was determined in a collection of cutaneous melanomas in the Thai population, implying that BRAF-targeted therapy may be a promising strategy for patients with BRAF-mutated cutaneous melanoma. This study revealed an association between the clinicopathological aspects of cutaneous melanoma and overall survival, disease-free survival, and overall mortality. A treatment with anti-BRAF-targeted therapy, which incorporates the already available medications' is being researched and developed.
Collapse
Affiliation(s)
- Jiraroch Meevassana
- From the Center of Excellence in Burn and Wound care, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Surawish Subbalekha
- From the Center of Excellence in Burn and Wound care, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tippawan Siritientong
- From the Center of Excellence in Burn and Wound care, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | | | - Pornthep Pungrasami
- From the Center of Excellence in Burn and Wound care, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kevin J Hamill
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Apichai Angsapatt
- From the Center of Excellence in Burn and Wound care, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
175
|
Kitrell BM, Blue ED, Siller A, Lobl MB, Evans TD, Whitley MJ, Wysong A. Gene Expression Profiles in Cutaneous Oncology. Dermatol Clin 2022; 41:89-99. [DOI: 10.1016/j.det.2022.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
176
|
Coit DG, Ariyan CE. Fifty years of progress in surgical oncology: Melanoma. J Surg Oncol 2022; 126:888-895. [PMID: 36087090 PMCID: PMC9473298 DOI: 10.1002/jso.27081] [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: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/07/2022]
Abstract
This paper outlines the scientific and clinical advances in the treatment of melanoma over the past 50 years. Among the highlights of progress, the dominant themes include evidence-based reduction in the extent and morbidity of surgical procedures in patients with local or regional melanoma without compromising end results, and the introduction of effective systemic therapy, specifically targeted therapy matched to patients based on specific tumor mutations, and immune checkpoint blockade. Management of advanced disease has also changed dramatically, due to improved understanding of the genomic variability of the disease as well as continuing improvements in imaging.
Collapse
Affiliation(s)
- Daniel G Coit
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Charlotte E Ariyan
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
177
|
|
178
|
Leong TW, McCook G, Frampton CM, Robinson BA, James ML. Adjuvant nodal field radiation in resected Stage III melanoma: A single-centre retrospective study in Christchurch, New Zealand. J Med Imaging Radiat Oncol 2022; 66:1003-1013. [PMID: 35642730 DOI: 10.1111/1754-9485.13438] [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: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Stage III melanoma is associated with poor outcomes. We studied the characteristics and outcomes of patients with resected Stage III melanoma before the routine use of adjuvant immunotherapy. Some of these patients received adjuvant nodal radiation with modern radiation techniques. METHODS We retrieved data of patients with resected Stage III melanoma treated in Christchurch over 10 years. Overall survival (OS), melanoma-specific survival (MSS), recurrence-free survival (RFS) and nodal recurrence-free rate (NRFR) were determined, and the association of these outcomes with tumour and treatment factors was investigated. RESULTS We identified 178 patients (110 male and 68 female), of whom 61 received adjuvant radiation. The median age was 66.6 years, and the median follow-up was 2.7 years. First recurrences occurred in 108 (61%) patients. There were 42 (24%) nodal field relapses and 103 (58%) distant relapses. One-half of nodal relapses in patients treated with adjuvant radiation were infield. The 5-year OS, RFS, MSS and NRFR were 46.4%, 26.8%, 53.7% and 69.6%, respectively. Adjuvant radiation was associated with improved RFS and no OS benefit. T4 disease and extranodal spread were associated with poorer OS, while extranodal spread and >3 involved nodes were associated with worse RFS. CONCLUSION Patients treated with adjuvant radiation remain at moderate risk of regional and high risk of distant relapse, despite the use of modern radiation techniques. Adjuvant radiation was associated with improved local control but infield recurrence rates remained a problem. The role of combined adjuvant radiation and immunotherapy in improving these outcomes requires further investigation.
Collapse
Affiliation(s)
- Tian Weng Leong
- Canterbury Regional Cancer and Haematology Service, Christchurch Hospital, Christchurch, New Zealand
| | - George McCook
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | | | - Bridget A Robinson
- Canterbury Regional Cancer and Haematology Service, Christchurch Hospital, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Melissa L James
- Canterbury Regional Cancer and Haematology Service, Christchurch Hospital, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
179
|
Abstract
The concept of precision medicine is based on the identification of hallmarks of cancer to exploit them as drug targets. The basic idea was that in this way the therapeutic modalities will be more effective and the side effects will be less. Since the majority of these novel modalities are not specific for a cancer-related biological process or a cancer-specific (mutant) target protein, it is not a surprise that we had to learn new type of side effects, because these therapeutics also affect physiological or pathological processes. Even more, in cases of some of these novel therapies we were able to discover new molecular mechanisms of physiological and pathological processes. Identification of the on-target side effects of targeted drugs can help to prevent the development of them or better manage the patients when emerge during cancer therapy.
Collapse
Affiliation(s)
- József Tímár
- Departments of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
- *Correspondence: József Tímár,
| | - Andrea Uhlyarik
- Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
180
|
Wall I, Boulat V, Shah A, Blenman KRM, Wu Y, Alberts E, Calado DP, Salgado R, Grigoriadis A. Leveraging the Dynamic Immune Environment Triad in Patients with Breast Cancer: Tumour, Lymph Node, and Peripheral Blood. Cancers (Basel) 2022; 14:4505. [PMID: 36139665 PMCID: PMC9496983 DOI: 10.3390/cancers14184505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022] Open
Abstract
During the anti-tumour response to breast cancer, the primary tumour, the peripheral blood, and the lymph nodes each play unique roles. Immunological features at each site reveal evidence of continuous immune cross-talk between them before, during and after treatment. As such, immune responses to breast cancer are found to be highly dynamic and truly systemic, integrating three distinct immune sites, complex cell-migration highways, as well as the temporal dimension of disease progression and treatment. In this review, we provide a connective summary of the dynamic immune environment triad of breast cancer. It is critical that future studies seek to establish dynamic immune profiles, constituting multiple sites, that capture the systemic immune response to breast cancer and define patient-selection parameters resulting in more significant overall responses and survival rates for breast cancer patients.
Collapse
Affiliation(s)
- Isobelle Wall
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Victoire Boulat
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Aekta Shah
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai 400012, India
| | - Kim R. M. Blenman
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Department of Computer Science, School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
| | - Yin Wu
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Elena Alberts
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Dinis Pedro Calado
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, 2610 Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| |
Collapse
|
181
|
Johannet P, Liu W, Fenyo D, Wind-Rotolo M, Krogsgaard M, Mehnert JM, Weber JS, Zhong J, Osman I. Baseline Serum Autoantibody Signatures Predict Recurrence and Toxicity in Melanoma Patients Receiving Adjuvant Immune Checkpoint Blockade. Clin Cancer Res 2022; 28:4121-4130. [PMID: 36106402 PMCID: PMC9662924 DOI: 10.1158/1078-0432.ccr-22-0404] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/02/2022] [Accepted: 05/18/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Adjuvant immunotherapy produces durable benefit for patients with resected melanoma, but many develop recurrence and/or immune-related adverse events (irAE). We investigated whether baseline serum autoantibody (autoAb) signatures predicted recurrence and severe toxicity in patients treated with adjuvant nivolumab, ipilimumab, or ipilimumab plus nivolumab. EXPERIMENTAL DESIGN This study included 950 patients: 565 from CheckMate 238 (408 ipilimumab versus 157 nivolumab) and 385 from CheckMate 915 (190 nivolumab versus 195 ipilimumab plus nivolumab). Serum autoAbs were profiled using the HuProt Human Proteome Microarray v4.0 (CDI Laboratories, Mayaguez, PR). Analysis of baseline differentially expressed autoAbs was followed by recurrence and severe toxicity signature building for each regimen, testing of the signatures, and additional independent validation for nivolumab using patients from CheckMate 915. RESULTS In the nivolumab independent validation cohort, high recurrence score predicted significantly worse recurrence-free survival [RFS; adjusted HR (aHR), 3.60; 95% confidence interval (CI), 1.98-6.55], and outperformed a model composed of clinical variables including PD-L1 expression (P < 0.001). Severe toxicity score was a significant predictor of severe irAEs (aHR, 13.53; 95% CI, 2.59-86.65). In the ipilimumab test cohort, high recurrence score was associated with significantly worse RFS (aHR, 3.21; 95% CI, 1.38-7.45) and severe toxicity score significantly predicted severe irAEs (aHR, 11.04; 95% CI, 3.84-37.25). In the ipilimumab plus nivolumab test cohort, high autoAb recurrence score was associated with significantly worse RFS (aHR, 6.45; 95% CI, 1.48-28.02), and high severe toxicity score was significantly associated with severe irAEs (aHR, 23.44; 95% CI, 4.10-212.50). CONCLUSIONS Baseline serum autoAb signatures predicted recurrence and severe toxicity in patients treated with adjuvant immunotherapy. Prospective testing of the signatures that include datasets with longer follow-up and rare but more severe toxicities will help determine their generalizability and potential clinical utility. See related commentary by Hassel and Luke, p. 3914.
Collapse
Affiliation(s)
- Paul Johannet
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Wenke Liu
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York
| | - David Fenyo
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York
| | | | - Michelle Krogsgaard
- Department of Pathology NYU, Grossman School of Medicine, New York, New York
| | - Janice M. Mehnert
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Jeffrey S. Weber
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Judy Zhong
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Iman Osman
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
182
|
Ulisse S, Baldini E, Pironi D, Gagliardi F, Tripodi D, Lauro A, Carbotta S, Tarroni D, D’Armiento M, Morrone A, Forte F, Frattaroli F, Persechino S, Odorisio T, D’Andrea V, Lori E, Sorrenti S. Is Melanoma Progression Affected by Thyroid Diseases? Int J Mol Sci 2022; 23:ijms231710036. [PMID: 36077430 PMCID: PMC9456309 DOI: 10.3390/ijms231710036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical and epidemiological evidence indicate a relationship between thyroid diseases and melanoma. In particular, the hypothyroidism condition appears to promote melanoma spread, which suggests a protective role of thyroid hormones against disease progression. In addition, experimental data suggest that, in addition to thyroid hormones, other hormonal players of the hypothalamic–pituitary–thyroid (HPT) axis, namely the thyrotropin releasing hormone and the thyrotropin, are likely to affect melanoma cells behavior. This information warrants further clinical and experimental studies in order to build a precise pattern of action of the HPT hormones on melanoma cells. An improved knowledge of the involved molecular mechanism(s) could lead to a better and possibly personalized clinical management of these patients.
Collapse
Affiliation(s)
- Salvatore Ulisse
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
- Correspondence:
| | - Enke Baldini
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Daniele Pironi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Federica Gagliardi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Domenico Tripodi
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Augusto Lauro
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Sabino Carbotta
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Danilo Tarroni
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Matteo D’Armiento
- Scientific Direction, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Aldo Morrone
- Scientific Direction, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Flavio Forte
- Urology Department, M.G. Vannini Hospital, 00177 Rome, Italy
| | - Flaminia Frattaroli
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Severino Persechino
- Department of Neurosciences, Mental Health and Sensory Organs, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Teresa Odorisio
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy
| | - Vito D’Andrea
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Eleonora Lori
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Salvatore Sorrenti
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| |
Collapse
|
183
|
Beisel C, Poretta T, Sheppard VB, Hurtado-de Mendoza A, Sipsma H, Fuqua E, Stwalley B, Salvatore A, Yang M. Adherence to Adjuvant Therapy in Patients with Resected Melanoma: An Application of the Theory of Planned Behavior. Adv Ther 2022; 39:4061-4075. [PMID: 35776398 PMCID: PMC9402483 DOI: 10.1007/s12325-022-02221-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Adherence to adjuvant therapy is crucial for effective disease management in patients with resected melanoma. This study assessed patient-reported adherence to adjuvant therapy and identified behavioral/belief constructs associated with adherence in patients with resected melanoma. METHODS Patients with resected stage III/IV melanoma were recruited through the Melanoma Research Foundation and a patient panel to complete an online survey. Patient characteristics, medical history, and adherence to therapy were captured. In accordance with the theory of planned behavior (TPB), the survey measured behavioral, normative, and control beliefs, and intention to adhere to therapy. Structural equation modeling (SEM) examined their relationships with adherence. RESULTS Among all patients who received adjuvant therapy and completed the survey (n = 184), 69% received intravenous and 31% received oral therapy; the majority (85.3%) were somewhat involved in deciding to start therapy. Mean age was 45 years, 44% of patients were female, and 83% had stage III/IV disease at diagnosis. Patients had a mean disease duration of 1.5 years, a time since complete resection of 10 months, and an adjuvant therapy duration of 8 months. Adherence to adjuvant therapy was high overall and higher with intravenous than with oral therapy (98.4% versus 91.2%, P = 0.002). All underlying TPB constructs were significant in the SEM model, which explained 60.3% of the variance in intention to adhere. Control beliefs had the strongest association with intention to adhere (standardized estimate = 0.47, P < 0.001) and intravenous therapy was associated with greater adherence than oral therapy (standardized estimate = 0.26, P < 0.001). CONCLUSIONS This study found that patients with resected melanoma are highly engaged in the decision to initiate systemic adjuvant therapy, with an overall high adherence rate to prescribed adjuvant treatment. Enhancing patients' view of their capability to adhere to treatments may further improve the adherence rate to melanoma adjuvant therapy.
Collapse
Affiliation(s)
| | - Tayla Poretta
- Bristol Myers Squibb, Princeton, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA.
| | - Vanessa B Sheppard
- Virginia Commonwealth University School of Medicine, Richmond and Virginia Commonwealth University Massey Cancer Center, Richmond, VA, USA
| | | | | | | | - Brian Stwalley
- Bristol Myers Squibb, Princeton, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Anthony Salvatore
- Bristol Myers Squibb, Princeton, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Min Yang
- Analysis Group, Inc., Boston, MA, USA
| |
Collapse
|
184
|
Garrison Z, Hornick N, Cheng J, Kulkarni RP. Circulating biomarkers of response to immunotherapy and immune-related adverse events. Expert Rev Mol Diagn 2022; 22:855-865. [PMID: 36193802 DOI: 10.1080/14737159.2022.2130688] [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: 01/08/2023]
Abstract
INTRODUCTION Immune checkpoint blockade has revolutionized cancer treatment. However, response rates vary, and these treatments have a high rate of immune-related side effects, which can be limiting. Thus, tests to predict who will respond and who may experience side effects are of critical importance toward realizing the ultimate goal of precision oncology. AREAS COVERED We review several of the most recent advances in circulating biomarkers that have been reported to be useful in predicting response and immune-related adverse events (irAE) to checkpoint blockade immunotherapies (CBI). We focus on high-quality studies published within the last few years. We highlight significant findings, identify areas for improvement, and provide recommendations on how these biomarkers may be translated into clinical utility. EXPERT OPINION As newer immunotherapies are developed, there is a pressing need to identify circulating biomarkers that can help predict responses and side effects. Current studies are mostly small-scale and retrospective; there is a need for larger-scale and prospective studies to help validate several of the biomarkers detailed here. As oncology focuses more on precision-based approaches, it is likely that a combination of biomarkers, including circulating ones as detailed here, will have critical utility in guiding clinical decisions.
Collapse
Affiliation(s)
- Zachary Garrison
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Noah Hornick
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey Cheng
- Department of Dermatology, University of California, San Francisco, CA, USA
| | - Rajan P Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.,Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.,Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
185
|
Adjuvant nivolumab versus ipilimumab (CheckMate 238 trial): Reassessment of 4-year efficacy outcomes in patients with stage III melanoma per AJCC-8 staging criteria. Eur J Cancer 2022; 173:285-296. [PMID: 35964471 DOI: 10.1016/j.ejca.2022.06.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/02/2022] [Accepted: 06/21/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE Nivolumab was approved as adjuvant therapy for melanoma based on data from CheckMate 238, which enrolled patients per American Joint Committee on Cancer version 7 (AJCC-7) criteria. Here, we analyse long-term outcomes per AJCC-8 staging criteria compared with AJCC-7 results to inform clinical decisions for patients diagnosed per AJCC-8. PATIENTS AND METHODS In a double-blind, phase 3 trial (NCT02388906), patients aged ≥15 years with resected, histologically confirmed AJCC-7 stage IIIB, IIIC, or IV melanoma were randomised to receive nivolumab 3 mg/kg every 2 weeks or ipilimumab 10 mg/kg every 3 weeks for 4 doses and then every 12 weeks, both intravenously ≤1 year. Recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) were assessed in patients with stage III disease, per AJCC-7 and AJCC-8. RESULTS Per AJCC-7 staging, 42.4% and 57.3% of patients were in substage IIIB and IIIC, respectively; per AJCC-8, 1.1%, 30.4%, 62.8%, and 5.0% were in IIIA, IIIB, IIIC, and IIID. After 4 years' minimum follow-up, the AJCC-7 superior efficacy of nivolumab over ipilimumab in patients with resected stage III melanoma was preserved per AJCC-8 analysis. No statistically significant difference in RFS between stage III substage hazard ratios was observed per AJCC-7 or -8 staging criteria (interaction test: AJCC-7, P = 0.8115; AJCC-8, P = 0.1051; P = 0.8392 ((AJCC-7) and P = 0.8678 (AJCC-8) for DMFS). CONCLUSIONS CheckMate 238 4-year RFS and DMFS outcomes are consistent per AJCC-7 and AJCC-8 staging criteria. Outcome benefits can therefore be translated for patients diagnosed per AJCC-8.
Collapse
|
186
|
Barricklow Z, DiVincenzo MJ, Angell CD, Carson WE. Ulcerated Cutaneous Melanoma: A Review of the Clinical, Histologic, and Molecular Features Associated with a Clinically Aggressive Histologic Phenotype. Clin Cosmet Investig Dermatol 2022; 15:1743-1757. [PMID: 36065342 PMCID: PMC9440663 DOI: 10.2147/ccid.s372287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/02/2022] [Indexed: 12/05/2022]
Abstract
The presence of ulceration in melanoma is associated with poor clinical outcomes and is the third most powerful predictor of survival in the AJCC Melanoma Staging System after tumor thickness and mitotic activity. The aggressive biological behavior associated with ulceration has been hypothesized to be the result of an intrinsic biological attribute that favors dissemination and presents locally with the loss of epidermal integrity. Among the features of ulcerated melanoma, many show promise as potential prognostic tools, markers of differential immunogenicity and indicators of oncogenic drivers of invasion and metastasis. The incidence of ulcerated melanoma is greater in males, increases with age and with systemic inflammatory risk factors (diabetes, smoking, low vitamin D, elevated body mass index). Patients with ulcerated primary tumors seem to exclusively benefit from adjuvant interferon (IFN) therapy, which is likely the consequence of an altered tumor microenvironment. When ulceration is present, there is a higher density of macrophages and dendritic cells and enhanced expression of pro-inflammatory cytokines, such as IL-6. There is also an increased expression of proteins involved in tumor antigen presentation in ulcerated melanomas. Histologically, vascular density, vasculogenic mimicry and angiotropism are all significantly correlated with ulceration in melanoma. The presence of ulceration is associated with reduced protein expression of E-cadherin and PTEN and elevated levels of N-cadherin and the matrix metalloproteinases. Differential microRNA expression also holds promise as a potential prognostic biomarker of malignancy and disease spread within the setting of ulceration. However, the molecular and cellular differences associated with the ulcerated state are complex and further study will aid in determining how these differences can be harnessed to improve care for patients with melanoma.
Collapse
Affiliation(s)
- Zoe Barricklow
- The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio, State University, Columbus, OH, USA
| | - Mallory J DiVincenzo
- The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio, State University, Columbus, OH, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Colin D Angell
- The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio, State University, Columbus, OH, USA
| | - William E Carson
- The Arthur G. James Cancer Hospital and Solove Research Institute, The Ohio, State University, Columbus, OH, USA
| |
Collapse
|
187
|
Treatment of Metastatic Melanoma at First Diagnosis: Review of the Literature. Life (Basel) 2022; 12:life12091302. [PMID: 36143339 PMCID: PMC9505710 DOI: 10.3390/life12091302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/27/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Metastatic melanoma (MM) is a pathological entity with a very poor prognosis that, until a few decades ago, had a low response rate to systemic treatments. Fortunately, in the last few years, new therapies for metastatic melanoma have emerged. Currently, targeted therapy and immunotherapy are the mainstays of the therapeutic arsenal available for patients with unresectable or metastatic melanoma. However, both clinical evolution and drug efficacy in melanoma patients are very different depending on the stage at which it is diagnosed. In fact, the aggressiveness of melanoma is different depending on whether it debuts directly as metastatic disease or if what occurs is a relapse after a first diagnosis at an early stage, although the biological determinants are largely unknown. Another key aspect in the clinical management of metastatic melanoma at first diagnosis strives in the different prognosis of melanoma of unknown primary (MUP) compared to melanoma of known primary (MPK). Understanding the mechanisms behind this, and the repercussion of implementing targeted and immune therapies in this specific form is crucial for designing diagnosis and treatment decision algorithms that optimize the current strategies. In this review article, we recapitulate the information available thus far regarding the epidemiology and response to immunotherapy treatments or targeted therapy in patients diagnosed with metastatic melanoma as a first diagnosis, with especial emphasis on the emerging specific information of the subpopulation formed by MUP patients.
Collapse
|
188
|
Panagi M, Pilavaki P, Constantinidou A, Stylianopoulos T. Immunotherapy in soft tissue and bone sarcoma: unraveling the barriers to effectiveness. Theranostics 2022; 12:6106-6129. [PMID: 36168619 PMCID: PMC9475460 DOI: 10.7150/thno.72800] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/21/2022] [Indexed: 11/05/2022] Open
Abstract
Sarcomas are uncommon malignancies of mesenchymal origin that can arise throughout the human lifespan, at any part of the body. Surgery remains the optimal treatment modality whilst response to conventional treatments, such as chemotherapy and radiation, is minimal. Immunotherapy has emerged as a novel approach to treat different cancer types but efficacy in soft tissue sarcoma and bone sarcoma is limited to distinct subtypes. Growing evidence shows that cancer-stroma cell interactions and their microenvironment play a key role in the effectiveness of immunotherapy. However, the pathophysiological and immunological properties of the sarcoma tumor microenvironment in relation to immunotherapy advances, has not been broadly reviewed. Here, we provide an up-to-date overview of the different immunotherapy modalities as potential treatments for sarcoma, identify barriers posed by the sarcoma microenvironment to immunotherapy, highlight their relevance for impeding effectiveness, and suggest mechanisms to overcome these barriers.
Collapse
Affiliation(s)
- Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | | | - Anastasia Constantinidou
- Medical School, University of Cyprus, Nicosia, Cyprus
- Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
189
|
Gao J, Miao J, Sun H, Fu X, Zhang P, Chen Z, Zhu P. TNF-α inhibitor ameliorates immune-related arthritis and pneumonitis in humanized mice. Front Immunol 2022; 13:955812. [PMID: 36016934 PMCID: PMC9396351 DOI: 10.3389/fimmu.2022.955812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThis study aimed at establishing a mouse model of immune-related adverse in humanized BALB/c-hPD1/hCTLA4 mice to investigate their potential pathogenesis and explore therapeutic targets for immune-related arthritis and pneumonitis.MethodsHumanized BALB/c-hPD1/hCTLA4 mice were injected with vehicle or collagen-specific antibodies (CA) and immune checkpoint inhibitors (ICI, ipilimumab, anti-human CTLA-4; and nivolumab, anti-human PD-1), and some mice were treated with anti-TNF-α antibody, leading to the control, collagen antibody-induced arthritis (CAIA), CAIA+ICI and treatment groups. The severity of clinical arthritis and pneumonitis in mice was monitored longitudinally and the pathological changes in the joints and lungs were histologically analyzed and the contents of lung hydroxyproline were measured. The frequency of different subsets of T cells was analyzed by flow cytometry and multiplex immunofluorescency.ResultsCompared with the control, the ICI group of mice developed the delayed onset of moderate degrees of arthritis while the CAIA+ICI group of mice exhibited the early onset of severe arthritis. Treatment with ICI caused severe pneumonitis, especially in the mice with CA. Flow cytometry analysis indicated a significantly higher frequency of splenic TNF-α+CD4+ and TNF-α+CD8+ T cells, but not other subsets of T cells tested, in the CAIA+ICI group of mice, relative to that in other groups of mice. Treatment with anti-TNF-α significantly mitigated the severity of arthritis and pneumonitis as well as deposition of collagen in lung of mice. The treatment also decreased the frequency of TNF-α+CD4+ and TNF-α+CD8+ T cells as well as effector memory T cells in the periphery lymph orangs and lungs of mice.ConclusionsWe successfully established a humanized mouse model of ICI-related severe arthritis and pneumonitis with a higher frequency of TNF-α+ T cells, which were significantly mitigated by anti-TNF-α treatment. Conceptually, ICI treatment can induce multiple autoimmune-like diseases in autoimmune-prone individuals and TNF-α+ T cells may be therapeutic targets for intervention of immune-related arthritis and pneumonitis.
Collapse
Affiliation(s)
- Jian Gao
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Jinlin Miao
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| | - Haoyang Sun
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xianghui Fu
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Peiyan Zhang
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhinan Chen
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| | - Ping Zhu
- Department of Clinical Immunology, National Translational Science Center for Molecular Medicine & Department of Cell Biology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Ping Zhu, ; Zhinan Chen, ; Jinlin Miao,
| |
Collapse
|
190
|
Kiss I, Kuhn M, Hrusak K, Buchler B, Boublikova L, Buchler T. Insomnia in patients treated with checkpoint inhibitors for cancer: A meta-analysis. Front Oncol 2022; 12:946307. [PMID: 35982959 PMCID: PMC9380599 DOI: 10.3389/fonc.2022.946307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeInsomnia in cancer patients is a common symptom contributing to poor quality of life and poor functioning. Sleep disturbances have been associated with inflammatory activity, and systemic cancer therapies chemotherapy, hormonal therapy, and immunotherapy may cause insomnia. We have carried out a meta-analysis to estimate the occurrence of insomnia in patients with solid cancer treated with immunotherapy using checkpoint inhibitors (CPI).MethodsPubMed and ClinicalTrials.gov were searched for phase 3 studies in solid tumours where treatment included a checkpoint inhibitor in the experimental arm. Data on the incidence of insomnia were acquired from the adverse events tables available from clinicaltrials.gov and/or from the full texts. Random effect logistic model was used to compare pooled data. Heterogeneity between studies was assessed using Cochrane Q statistics and I2 statistics.ResultsA total of 54 studies (including six three-arm studies) involving 37,352 patients were included in the analysis. Insomnia was reported in 8.3% of subjects (95% confidence interval [CI] 8.0%-8.7%) treated with immunotherapy. Insomnia was significantly more common in patients receiving immunotherapy compared to those enrolled in study arms with inactive treatment (odds ratio [OR] 1.49, 95% CI 1.13-1.96). The odds for insomnia were similar between the arms for studies comparing CPI versus chemotherapy and CPI versus non-immunologic targeted therapies (OR 1.07, 95% CI 0.94-1.22 and OR 1.40, 95% CI 0.90-2.18, respectively). The OR for insomnia was higher for cytotoxic T-lymphocyte antigen 4 (CTLA-4) receptor inhibitors compared to the inhibitors of programmed death-1 (PD-1) receptor (OR 1.36, 95% CI 1.06 – 1.74).ConclusionCancer immunotherapy using CPI is associated with insomnia but the odds of developing the symptom are not greater with immunotherapy than with other systemic modalities including chemotherapy and non-immunologic targeted therapies.
Collapse
Affiliation(s)
- Igor Kiss
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Matyas Kuhn
- Institute of Biostatistics and Analyses, Masaryk University, Brno, Czechia
| | - Kristian Hrusak
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Benjamin Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Ludmila Boublikova
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
- *Correspondence: Tomas Buchler,
| |
Collapse
|
191
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
192
|
Lao Y, Shen D, Zhang W, He R, Jiang M. Immune Checkpoint Inhibitors in Cancer Therapy—How to Overcome Drug Resistance? Cancers (Basel) 2022; 14:cancers14153575. [PMID: 35892835 PMCID: PMC9331941 DOI: 10.3390/cancers14153575] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Immune checkpoint inhibitors (ICIs) are an important strategy in cancer therapy. However, with the widespread clinical use of ICIs, people gradually found that ICIs may not be effective enough to eliminate tumor tissue for certain patients. The resistance to ICI treatment makes some patients unable to benefit from their antitumor effects. Therefore, it is vital to understand their antitumor and drug resistance mechanisms to better narrow the ICI-resistant patient population. This review outlines the antitumor action sites and mechanisms of different types of ICIs and lists the main reason of ICI resistance based on recent studies. Finally, we propose current and future solutions for resistance to ICIs. Abstract Immune checkpoint inhibitors (ICIs), antagonists used to remove tumor suppression of immune cells, have been widely used in clinical settings. Their high antitumor effect makes them crucial for treating cancer after surgery, radiotherapy, chemotherapy, and targeted therapy. However, with the advent of ICIs and their use by a large number of patients, more clinical data have gradually shown that some cancer patients still have resistance to ICI treatment, which makes some patients unable to benefit from their antitumor effect. Therefore, it is vital to understand their antitumor and drug resistance mechanisms. In this review, we focused on the antitumor action sites and mechanisms of different types of ICIs. We then listed the main possible mechanisms of ICI resistance based on recent studies. Finally, we proposed current and future solutions for the resistance of ICIs, providing theoretical support for improving their clinical antitumor effect.
Collapse
Affiliation(s)
- Yefang Lao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China;
| | - Daoming Shen
- Department of Internal Medicine, Xiangcheng People’s Hospital, Suzhou 215131, China;
| | - Weili Zhang
- Department of Gastroenterology, Xiangcheng People’s Hospital, Suzhou 215131, China;
| | - Rui He
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, Shanghai 200433, China
- Correspondence: (R.H.); (M.J.); Tel.: +86-18862185684 (R.H.); +86-13776022109 (M.J.)
| | - Min Jiang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China;
- Correspondence: (R.H.); (M.J.); Tel.: +86-18862185684 (R.H.); +86-13776022109 (M.J.)
| |
Collapse
|
193
|
Yu Y, Zhou Y, Zhang X, Tan K, Zheng J, Li J, Cui H. Immune Checkpoint Inhibitors in the Treatment of Patients With Cancer and Preexisting Psoriasis: A Systematic Review and Meta-Analysis of Observational Studies. Front Oncol 2022; 12:934093. [PMID: 35912183 PMCID: PMC9334704 DOI: 10.3389/fonc.2022.934093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Immunotherapies represented by immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. A large part of the population has both cancer and psoriasis but is usually excluded from ICI clinical trials because of the dysregulated activation of the immune system. This is the first study to evaluate the safety and efficacy of ICI therapy in patients with cancer and preexisting psoriasis. Methods PubMed, EMBASE, Cochrane, and MEDLINE databases were searched from inception through February 2022. Observational studies on patients with cancer and confirmed psoriasis before ICI initiation were included. Outcomes included the incidence of psoriasis flares, de novo immune-related adverse events (irAEs), discontinuation rate due to flare/de novo irAEs, and efficacy of ICI therapy. Clinical manifestations, management, and outcomes for adverse events (AEs) were systematically reviewed. All pooled analyses were based on a random-effects model using Stata software. Meta-regression and subgroup analyses were performed to identify sources of heterogeneity. Results Twelve studies involving 191 patients were included. The pooled incidence of psoriasis flares was 45.0% (95% CI: 31.1%-58.9%, I2 = 71.7%) and 44.9% (95% CI: 29.0%–60.7%, I2 = 71.8%) for de novo irAEs. The tumor type, psoriasis subtype, ICI class, and country were the main sources of heterogeneity. Grade 3–4 flares occurred in 10.8% (95% CI: 5.3%–16.3%) of patients, and about 16.6% (95% CI: 10.7%–22.5%) of patients experienced grade 3–4 de novo irAEs. The estimated incidence of ICI discontinuation due to AE was 18.5% (95% CI: 6.1%–30.8%, I2 = 68.7%). The median times to develop flare and de novo irAEs were 44 and 63 days, respectively. Endocrinopathies and colitis were the most common de novo irAEs. Conventional therapy is effective for most AEs. The estimated objective response rate (ORR) of ICIs was 38.1% (95% CI: 11.8%–64.3%, I2 = 81.7%), and the disease control rate (DCR) was 64.5% (95% CI: 55.3%–73.8%, I2 = 0). Conclusions The flare of patients with cancer and preexisting psoriasis treated with ICI therapy is frequent, but the incidence of de novo irAEs and the efficacy of ICI therapy are comparable to those of the general population. Most AEs are mild and manageable with conventional therapy, which required discontinuation of ICI therapy in 18.5%. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022320646
Collapse
Affiliation(s)
- Yixuan Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yang Zhou
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Kexin Tan
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jiabin Zheng
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jia Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Huijuan Cui
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Huijuan Cui,
| |
Collapse
|
194
|
Druce I, Tawagi K, Shaw JLV, Ibrahim A, Lochnan H, Ong M. Routine Screening for Central and Primary Adrenal Insufficiency during Immune-Checkpoint Inhibitor Therapy: An Endocrinology Perspective for Oncologists. Curr Oncol 2022; 29:4665-4677. [PMID: 35877230 PMCID: PMC9315594 DOI: 10.3390/curroncol29070370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Immune checkpoint inhibitor (ICI)-associated hypothalamic–pituitary–adrenal axis disruption can lead to hypocortisolism. This is a life-threatening but difficult to diagnose condition, due to its non-specific symptoms that overlap with symptoms of malignancy. Currently, there is no consensus on how to best screen asymptomatic patients on ICI therapy for hypophysitis with serum cortisol. Methods: A retrospective chart review of patients treated with ICI in a tertiary care centre was conducted to assess the rate of screening with cortisol and whether this had an impact on diagnosis of ICI-hypophysitis in the preclinical stage. Patients were identified as having hypophysitis with an adrenocorticotropin hormone (ACTH) deficiency based on chart review of patients with cortisol values ≤ 140 nmol/L (≤5 mcg/dL). We also assessed what proportion of cortisol values were drawn at the correct time for interpretation (between 6 AM and 10 AM). Results: Two hundred and sixty-five patients had 1301 cortisol levels drawn, only 40% of which were drawn correctly (between 6 and 10 AM). Twenty-two cases of hypophysitis manifesting with ACTH deficiency were identified. Eight of these patients were being screened with cortisol following treatment and were detected in the outpatient setting. The remaining 14 patients were not screened and were diagnosed when symptomatic, after an emergency room visit or hospital admission. Sixty percent of the cortisol tests were uninterpretable as they were not drawn within the appropriate time window. Conclusion: Measuring morning serum cortisol in asymptomatic patients on ICI therapy is a fast and inexpensive way to screen for hypophysitis and should become the standard of care. Random serum cortisol measurement has no clinical value. Education needs to be provided on when to correctly perform the test and how to interpret it and we provide an algorithm for this purpose. The adoption and validation of such an algorithm as part of routine practice could significantly reduce morbidity and mortality in patients, especially as ICI therapy is becoming increasingly commonplace.
Collapse
Affiliation(s)
- Irena Druce
- Division of Endocrinology and Metabolism, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON KIH8L6, Canada;
- Correspondence:
| | - Karine Tawagi
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON KIH8L6, Canada; (K.T.); (A.I.); (M.O.)
| | - Julie L. V. Shaw
- Department of Pathology and Laboratory Medicine, Eastern Ontario Regional Laboratories Association, The Ottawa Hospital Research Institute, The Ottawa Hospital, The University of Ottawa, Ottawa, ON KIH8L6, Canada;
| | - Andrea Ibrahim
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON KIH8L6, Canada; (K.T.); (A.I.); (M.O.)
| | - Heather Lochnan
- Division of Endocrinology and Metabolism, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON KIH8L6, Canada;
| | - Michael Ong
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON KIH8L6, Canada; (K.T.); (A.I.); (M.O.)
| |
Collapse
|
195
|
Eggermont AMM, Hamid O, Long GV, Luke JJ. Optimal systemic therapy for high-risk resectable melanoma. Nat Rev Clin Oncol 2022; 19:431-439. [PMID: 35468949 PMCID: PMC11075933 DOI: 10.1038/s41571-022-00630-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Immunotherapy with immune-checkpoint inhibitors and molecularly targeted therapy with BRAF inhibitors were pioneered in the setting of advanced-stage, unresectable melanoma, where they revolutionized treatment and considerably improved patient survival. These therapeutic approaches have also been successfully transitioned into the resectable disease setting, with the regulatory approvals of ipilimumab, pembrolizumab, nivolumab, and dabrafenib plus trametinib as postoperative (adjuvant) treatments for various, overlapping groups of patients with high-risk melanoma. Moreover, these agents have shown variable promise when used in the preoperative (neoadjuvant) period. The expanding range of treatment options available for resectable high-risk melanoma, all of which come with risks as well as benefits, raises questions over selection of the optimal therapeutic strategy and agents for each individual, also considering that many patients might be cured with surgery alone. Furthermore, the use of perioperative therapy has potentially important implications for the management of patients who have disease recurrence. In this Viewpoint, we asked four expert investigators and medical or surgical oncologists who have been involved in the key studies of perioperative systemic therapies for their perspectives on the optimal management of patients with high-risk melanoma.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- Comprehensive Cancer Center Munich, Munich, Germany.
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands.
- University Medical Center Utrecht, Utrecht, Netherlands.
| | - Omid Hamid
- The Angeles Clinic and Research Institute, Cedar Sinai Affiliate, Los Angeles, CA, USA.
| | - Georgia V Long
- Melanoma Institute Australia and Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Mater and Royal North Shore Hospitals, Sydney, New South Wales, Australia.
| | - Jason J Luke
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
196
|
Zamagni F, Bucchi L, Mancini S, Crocetti E, Dal Maso L, Ferretti S, Biggeri A, Villani S, Baldacchini F, Giuliani O, Ravaioli A, Vattiato R, Brustolin A, Candela G, Carone S, Carrozzi G, Cavallo R, Dinaro YM, Ferrante M, Iacovacci S, Mazzoleni G, Musolino A, Rizzello RV, Serraino D, Stracci F, Tumino R, Masini C, Ridolfi L, Palmieri G, Stanganelli I, Falcini F, the AIRTUM Working Group. The relative contribution of the decreasing trend in tumour thickness to the 2010s increase in net survival from cutaneous malignant melanoma in Italy: a population-based investigation. Br J Dermatol 2022; 187:52-63. [PMID: 35253204 PMCID: PMC9542017 DOI: 10.1111/bjd.21051] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND The long-term increase in survival from cutaneous malignant melanoma (CMM) is generally attributed to the decreasing trend in tumour thickness, the single most important prognostic factor. OBJECTIVES To determine the relative contribution of decreased tumour thickness to the favourable trend in survival from CMM in Italy. METHODS Eleven local cancer registries covering a population of 8 056 608 (13.4% of the Italian population in 2010) provided records for people with primary CMM registered between 2003 and 2017. Age-standardized 5-year net survival was calculated. Multivariate analysis of 5-year net survival was undertaken by calculating the relative excess risk (RER) of death. The relative contribution of the decrease in tumour thickness to the RER of death was evaluated using a forward stepwise flexible parametric survival model including the available prognostic factors. RESULTS Over the study period, tumour thickness was inversely associated with 5-year net survival and multivariate RER in both sexes. The median thickness was 0.90 mm in 2003-2007, 0.85 mm in 2008-2012 and 0.75 mm in 2013-2017 among male patients, and 0.78 mm, 0.77 mm and 0.68 mm among female patients, respectively. The 5-year net survival was 86.8%, 89.2% and 93.2% in male patients, and 91.4%, 92.0% and 93.4% in female patients, respectively. In 2013-2017, male patients exhibited the same survival as female patients despite having thicker lesions. For them, the increasing survival trend was more pronounced with increasing thickness, and the inclusion of thickness in the forward stepwise model made the RER in 2013-2017 vs. 2003-2007 increase from 0.64 [95% confidence interval (CI) 0.51-0.80] to 0.70 (95% CI 0.57-0.86). This indicates that the thickness trend accounted for less than 20% of the survival increase. For female patients, the results were not significant but, with multiple imputation of missing thickness values, the RER rose from 0.74 (95% CI 0.58-0.93) to 0.82 (95% CI 0.66-1.02) in 2013-2017. CONCLUSIONS For male patients in particular, decrease in tumour thickness accounted for a small part of the improvement in survival observed in 2013-2017. The introduction of targeted therapies and immune checkpoint inhibitors in 2013 is most likely to account for the remaining improvement.
Collapse
Affiliation(s)
- Federica Zamagni
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Lauro Bucchi
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Silvia Mancini
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Emanuele Crocetti
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Luigino Dal Maso
- Cancer Epidemiology UnitCentro di Riferimento Oncologico di Aviano (CRO) IRCCSAvianoItaly
| | - Stefano Ferretti
- Romagna Cancer Registry, Section of FerraraLocal Health Authority and University of FerraraFerraraItaly
| | - Annibale Biggeri
- Department of Statistics, Computer Science, Applications G. ParentiUniversity of FlorenceFlorenceItaly
| | - Simona Villani
- Department of Public Health, Experimental and Forensic Medicine, Unit of Biostatistics and Clinical EpidemiologyUniversity of PaviaPaviaItaly
| | - Flavia Baldacchini
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Orietta Giuliani
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Alessandra Ravaioli
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Rosa Vattiato
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Angelita Brustolin
- UOSD Epidemiologia e Registro Tumori (Dip. di Prevenzione ASL VT) c/o Cittadella della SaluteViterboItaly
| | - Giuseppa Candela
- Trapani Cancer Registry, Dipartimento di Prevenzione della SaluteServizio Sanitario Regionale Sicilia, Azienda Sanitaria Provinciale (ASP)TrapaniItaly
| | - Simona Carone
- Registro tumori di TarantoUnità operativa complessa di statistica ed epidemiologiaAzienda sanitaria locale TarantoItaly
| | - Giuliano Carrozzi
- Modena Cancer Registry, Public Health DepartmentLocal Health AuthorityModenaItaly
| | | | | | - Margherita Ferrante
- Integrated Cancer Registry of Catania‐Messina‐EnnaAzienda Ospedaliero‐Universitaria Policlinico ‘Rodolico‐San Marco’CataniaItaly
| | | | | | - Antonino Musolino
- Department of Medicine and SurgeryUniversity of Parma; Medical Oncology Unit and Cancer Registry, University Hospital of ParmaParmaItaly
| | - Roberto Vito Rizzello
- Trento Province Cancer Registry, Unit of Clinical EpidemiologyAzienda Provinciale per i Servizi Sanitari (APSS) TrentoItaly
| | - Diego Serraino
- Cancer Epidemiology UnitCentro di Riferimento Oncologico di Aviano (CRO) IRCCSAvianoItaly
| | - Fabrizio Stracci
- Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Rosario Tumino
- Former Director Cancer RegistryProvincial Health Authority (ASP)RagusaItaly
| | - Carla Masini
- Unit of Oncological PharmacyIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Laura Ridolfi
- Immunotherapy, Cell Therapy and BiobankIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
| | - Giuseppe Palmieri
- Institute of Research on Genetics and Biomedicine (IRGB), National Research Council (CNR)SardegnaSassariItaly
| | - Ignazio Stanganelli
- Skin Cancer UnitIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
- Department of DermatologyUniversity of ParmaParmaItaly
| | - Fabio Falcini
- Romagna Cancer RegistryIRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) ‘Dino Amadori’MeldolaForlìItaly
- Cancer Prevention UnitLocal Health AuthorityForlìItaly
| | | |
Collapse
|
197
|
Franke V, Stahlie EHA, van der Hiel B, van de Wiel BA, Wouters MWJM, van Houdt WJ, van Akkooi ACJ. Re-introduction of T-VEC Monotherapy in Recurrent Melanoma is Effective. J Immunother 2022; 45:263-266. [PMID: 35580326 DOI: 10.1097/cji.0000000000000423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 04/14/2022] [Indexed: 11/26/2022]
Abstract
Talimogene laherparepvec (T-VEC) is a modified herpes simplex virus type 1, which can be administered intralesionally in patients with stage IIIB/C-IVM1a (American Joint Committee of Cancer; AJCC 7th edition) unresectable melanoma. In the case of disease recurrence, T-VEC can be re-introduced for the same category of patients. Five patients with recurrent disease after a prior achieved complete response (CR) recommenced treatment with T-VEC monotherapy at the Netherlands Cancer Institute. We collected data on response, adverse events and baseline characteristics. All 5 patients that were re-treated with T-VEC presented with in-transit metastases on the lower limb. Median age at baseline was 72.1 years with a median follow-up time of 30.4 months. Histologically proven CR was achieved after a median of 8 T-VEC courses on the initial exposure. Duration of response (time between first CR and recurrence) varied between 3.8 and 14.2 months. All 5 patients achieved a histologically and/or positron emission tomography/computed tomography proven CR again after re-introduction of T-VEC with a median of 5 courses. One patient (20%) developed a second recurrence and is currently still on treatment with T-VEC. No patients developed distant metastases. Grade 1 adverse events occurred in all patients. Mostly, these consisted of fatigue, influenza-like symptoms and injection site pain. Response to re-introduction of T-VEC monotherapy in this select patient population is promising. This real world data on re-introduction of T-VEC monotherapy in stage IIIB/C-IVM1a melanoma suggests T-VEC could be a treatment option for chronic disease control.
Collapse
Affiliation(s)
| | | | | | - Bart A van de Wiel
- Pathology at the Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | | | - Alexander C J van Akkooi
- Departments of Surgical Oncology
- Department of Surgical Oncology/Faculty Member Melanoma Institute Australia, The Poche Centre, Australia
| |
Collapse
|
198
|
Garbe C, Amaral T, Peris K, Hauschild A, Arenberger P, Basset-Seguin N, Bastholt L, Bataille V, Del Marmol V, Dréno B, Fargnoli MC, Forsea AM, Grob JJ, Hoeller C, Kaufmann R, Kelleners-Smeets N, Lallas A, Lebbé C, Lytvynenko B, Malvehy J, Moreno-Ramirez D, Nathan P, Pellacani G, Saiag P, Stratigos AJ, Van Akkooi ACJ, Vieira R, Zalaudek I, Lorigan P. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment - Update 2022. Eur J Cancer 2022; 170:256-284. [PMID: 35623961 DOI: 10.1016/j.ejca.2022.04.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A unique collaboration of multidisciplinary experts from the European Dermatology Forum (EDF), the European Association of Dermato-Oncology (EADO), and the European Organization of Research and Treatment of Cancer (EORTC) was formed to make recommendations on cutaneous melanoma diagnosis and treatment, based on the systematic literature reviews and the experts' experience. Cutaneous melanomas are excised with one to 2-cm safety margins. Sentinel lymph node dissection shall be performed as a staging procedure in patients with tumor thickness ≥1.0 mm or ≥0.8 mm with additional histological risk factors, although there is as yet no clear survival benefit for this approach. Therapeutic decisions in stage III/IV patients should be primarily made by an interdisciplinary oncology team ("tumor board"). Adjuvant therapies can be proposed in stage III/completely resected stage IV patients and are primarily anti-PD-1, independent of mutational status, or alternatively dabrafenib plus trametinib for BRAF mutant patients. In distant metastases (stage IV), either resected or not, systemic treatment is always indicated. For first-line treatment particularly in BRAF wild-type patients, immunotherapy with PD-1 antibodies alone or in combination with CTLA-4 antibodies shall be considered. In stage IV melanoma with a BRAF-V600 E/K mutation, first-line therapy with BRAF/MEK inhibitors can be offered as an alternative to immunotherapy. In patients with primary resistance to immunotherapy and harboring a BRAF-V600 E/K mutation, this therapy shall be offered as second-line therapy. Systemic therapy in stage III/IV melanoma is a rapidly changing landscape, and it is likely that these recommendations may change in the near future.
Collapse
Affiliation(s)
- Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany.
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Ketty Peris
- Institute of Dermatology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Petr Arenberger
- Department of Dermatovenereology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicole Basset-Seguin
- Université Paris Cite, AP-HP, Department of Dermatology INSERM U 976 Hôpital, Saint Louis Paris France
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Veronique Bataille
- Twin Research and Genetic Epidemiology Unit, School of Basic & Medical Biosciences, King's College London, London, SE1 7EH, United Kingdom
| | - Veronique Del Marmol
- Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Brigitte Dréno
- Dermatology Department, CHU Nantes, CIC 1413, CRCINA, University Nantes, Nantes, France
| | - Maria C Fargnoli
- Dermatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Ana-Maria Forsea
- Dermatology Department, Elias University Hospital, Carol Davila University of Medicine and Pharmacy Bucharest, Romania
| | | | | | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology, Frankfurt University Hospital, Frankfurt, Germany
| | | | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Celeste Lebbé
- Université Paris Cite, AP-HP, Department of Dermatology INSERM U 976 Hôpital, Saint Louis Paris France
| | - Bodhan Lytvynenko
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Josep Malvehy
- Melanoma Unit, Department of Dermatology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - David Moreno-Ramirez
- Medical-&-Surgical Dermatology Service, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Paul Nathan
- Mount-Vernon Cancer Centre, Northwood United Kingdom
| | | | - Philippe Saiag
- University Department of Dermatology, Université de Versailles-Saint Quentin en Yvelines, APHP, Boulogne, France
| | - Alexander J Stratigos
- First Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Alexander C J Van Akkooi
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Ricardo Vieira
- Department of Dermatology and Venereology, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Iris Zalaudek
- Dermatology Clinic, Maggiore Hospital, University of Trieste, Trieste, Italy
| | - Paul Lorigan
- The University of Manchester, Oxford Rd, Manchester, M13 9PL, United Kingdom
| |
Collapse
|
199
|
O'Quigley J. Testing for Differences in Survival When Treatment Effects Are Persistent, Decaying, or Delayed. J Clin Oncol 2022; 40:3537-3545. [PMID: 35767775 DOI: 10.1200/jco.21.01811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A statistical test for the presence of treatment effects on survival will be based on a null hypothesis (absence of effects) and an alternative (presence of effects). The null is very simply expressed. The most common alternative, also simply expressed, is that of proportional hazards. For this situation, not only do we have a very powerful test in the log-rank test but also the outcome is readily interpreted. However, many modern treatments fall outside this relatively straightforward paradigm and, as such, have attracted attention from statisticians eager to do their best to avoid losing power as well as to maintain interpretability when the alternative hypothesis is less simple. Examples include trials where the treatment effect decays with time, immunotherapy trials where treatment effects may be slow to manifest themselves as well as the so-called crossing hazards problem. We review some of the solutions that have been proposed to deal with these issues. We pay particular attention to the integrated log-rank test and how it can be combined with the log-rank test itself to obtain powerful tests for these more complex situations.
Collapse
Affiliation(s)
- John O'Quigley
- Department of Statistical Science, University College London, London, United Kingdom
| |
Collapse
|
200
|
Jiang M, Hu Y, Lin G, Chen C. Dosing Regimens of Immune Checkpoint Inhibitors: Attempts at Lower Dose, Less Frequency, Shorter Course. Front Oncol 2022; 12:906251. [PMID: 35795044 PMCID: PMC9251517 DOI: 10.3389/fonc.2022.906251] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/24/2022] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a revolutionary breakthrough in the field of cancer by modulating patient's own immune system to exert anti-tumor effects. The clinical application of ICIs is still in its infancy, and their dosing regimens need to be continuously adjusted. Pharmacokinetic/pharmacodynamic studies showed a significant plateau in the exposure-response curve, with high receptor occupancy and plasma concentrations achieved at low dose levels. Coupled with concerns about drug toxicity and heavy economic costs, there has been an ongoing quest to reevaluate the current ICI dosing regimens while preserving maximum clinical efficacy. Many clinical data showed remarkable anticancer effects with ICIs at the doses far below the approved regimens, indicating the possibility of dose reduction. Our review attempts to summarize the clinical evidence for ICIs regimens with lower-dose, less-frequency, shorter-course, and provide clues for further ICIs regimen optimization.
Collapse
Affiliation(s)
| | | | | | - Chao Chen
- Department of Radiotherapy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|