151
|
Fukushima S, Miyashita A, Kimura T, Kuriyama H, Mizuhashi S, Ichigozaki Y, Masuguchi S. Deciphering the role of adjuvant therapy in melanoma and its actual benefits. J Dermatol 2024; 51:335-342. [PMID: 38212945 PMCID: PMC11483963 DOI: 10.1111/1346-8138.17093] [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: 11/17/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024]
Abstract
Numerous clinical trials have demonstrated a significant improvement in recurrence-free survival among melanoma patients receiving high-dose interferon-α, immune checkpoint inhibitors (pembrolizumab, nivolumab), and BRAF/MEK inhibitors (dabrafenib-trametinib). This study aimed to investigate whether these findings hold true in real-world conditions for patients with stage III and IV melanoma. In particular, the study explores the efficacy and side effects of adjuvant therapies, focusing on anti-PD-1 antibodies and BRAF/MEK inhibitors. While clinical trials have shown comparable efficacy, differences in side-effect profiles, especially the persistence of immune-related adverse events with anti-PD-1 antibodies, highlight the need for careful consideration in adjuvant settings. In the absence of established biomarkers for guiding adjuvant therapy decisions, it becomes imperative to transparently communicate the advantages and disadvantages of drug administration to patients. The study also delved into the impact of melanoma subtype and BRAF mutation status on the effectiveness of adjuvant therapy, emphasizing the need for further investigation.
Collapse
Affiliation(s)
- Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Toshihiro Kimura
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Haruka Kuriyama
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Satoru Mizuhashi
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Yuki Ichigozaki
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Shinichi Masuguchi
- Department of Dermatology and Plastic Surgery, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
152
|
Rutkowski P, Mandala M. Perioperative therapy of melanoma: Adjuvant or neoadjuvant treatment. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:107969. [PMID: 38342039 DOI: 10.1016/j.ejso.2024.107969] [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: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 02/13/2024]
Abstract
Surgery is the mainstay treatment of melanoma. However, even after radical resection the risk of relapses in majority of stage IIB-IV disease remains high. Currently, the standard treatment after surgery in high risk patients is systemic adjuvant therapy administered up to one year based on the results of clinical trials indicating significant reduction of risk of relapses. All clinical trials in adjuvant setting were based as primary end-point on relapse-free survival, not overall survival, and they did not incorporate and validate biomarkers prospectively. A new therapeutic strategy in locoregional advanced melanomas becomes a preoperative treatment to further increase of the cure rates and decrease the duration of systemic therapy.
Collapse
Affiliation(s)
- Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Mario Mandala
- University of Perugia, Santa Maria Misericordia Hospital, Piazza Menghini 1, 06132, Perugia, Italy
| |
Collapse
|
153
|
Mattila KE, Vihinen H, Heervä E, Nuotio MS, Vihinen P. The impact of prognostic factors and comorbidities on survival in older adults with stage I - III cutaneous melanoma in Southwest Finland: A register study. J Geriatr Oncol 2024; 15:101701. [PMID: 38219332 DOI: 10.1016/j.jgo.2023.101701] [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: 09/13/2023] [Revised: 11/23/2023] [Accepted: 12/22/2023] [Indexed: 01/16/2024]
Abstract
INTRODUCTION Despite being diagnosed with thicker and more often ulcerated melanomas, cancer-specific survival (CSS) is not necessarily inferior in older adults with melanoma compared to younger patients. MATERIALS AND METHODS Our aim was to evaluate the impact of baseline melanoma-specific prognostic factors and comorbidities on recurrence-free survival (RFS), CSS, and overall survival (OS) in patients aged 70-79 (n = 474) and ≥ 80 years (n = 286) with resected stage I - III cutaneous melanoma in Southwest Finland between January 1, 2000 and December 31, 2020. Patients were restaged according to the 8th edition of TNM classification, and comorbidities were assessed using the Charlson Comorbidity Index (CCI). RESULTS Patients aged ≥80 years had thicker and more commonly ulcerated melanomas: 43.0%, 40.9%, and 16.1% of patients aged ≥80 and 56.5%, 25.3%, and 18.1% of patients aged 70-79 years were diagnosed with stage I, II, and III melanoma, respectively. Multiple comorbidities (CCI ≥2) were more common and sentinel lymph node biopsy less frequently performed in patients aged ≥80 years. RFS and CSS were similar in patients aged 70-79 years and ≥ 80 years: median RFS 13.8 years vs not reached, with the hazard ratio of melanoma recurrence or death from melanoma 1.25 (95% confidence interval [CI]: 0.91-1.71); median CSS was not reached, with the hazard ratio of death from melanoma 1.12 (95%CI: 0.81-1.75). The proportion of patients who were alive with melanoma recurrence or had died from melanoma was similar in both age groups. In multivariable analysis, higher pathological stage was the only independent risk factor for short RFS regardless of age group, sex, CCI, and tumor ulceration. Higher stage and male sex were associated with short CSS. Age ≥ 80 years, stage III disease, and CCI ≥ 2 were associated with short OS and female sex with long OS in multivariable analysis. DISCUSSION Pathological stage was the most influential factor determining RFS and CSS in older adults with resected stage I - III melanoma. Concerning OS, age ≥ 80 years, stage III disease, and multiple comorbidities had a significant negative impact.
Collapse
Affiliation(s)
- Kalle E Mattila
- Department of Oncology, Fican West Cancer Centre, University of Turku and Turku University Hospital, Finland; InFLAMES Research Flagship Center, University of Turku, Finland.
| | - Helmi Vihinen
- Department of Oncology, Fican West Cancer Centre, University of Turku and Turku University Hospital, Finland; Turku School of Economics, University of Turku, Finland
| | - Eetu Heervä
- InFLAMES Research Flagship Center, University of Turku, Finland
| | - Maria S Nuotio
- Department of Geriatric Medicine, University of Turku and Turku University Hospital, Finland
| | - Pia Vihinen
- Department of Oncology, Fican West Cancer Centre, University of Turku and Turku University Hospital, Finland
| |
Collapse
|
154
|
Olofsson Bagge R, Mikiver R, Marchetti MA, Lo SN, van Akkooi ACJ, Coit DG, Ingvar C, Isaksson K, Scolyer RA, Thompson JF, Varey AHR, Wong SL, Lyth J, Bartlett EK. Population-Based Validation of the MIA and MSKCC Tools for Predicting Sentinel Lymph Node Status. JAMA Surg 2024; 159:260-268. [PMID: 38198163 PMCID: PMC10782377 DOI: 10.1001/jamasurg.2023.6904] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/13/2023] [Indexed: 01/11/2024]
Abstract
Importance Patients with melanoma are selected for sentinel lymph node biopsy (SLNB) based on their risk of a positive SLN. To improve selection, the Memorial Sloan Kettering Cancer Center (MSKCC) and Melanoma Institute Australia (MIA) developed predictive models, but the utility of these models remains to be tested. Objective To determine the clinical utility of the MIA and MSKCC models. Design, Setting, and Participants This was a population-based comparative effectiveness research study including 10 089 consecutive patients with cutaneous melanoma undergoing SLNB from the Swedish Melanoma Registry from January 2007 to December 2021. Data were analyzed from May to August 2023. Main Outcomes and Measures, The predicted probability of SLN positivity was calculated using the MSKCC model and a limited MIA model (using mitotic rate as absent/present instead of count/mm2 and excluding the optional variable lymphovascular invasion) for each patient. The operating characteristics of the models were assessed and compared. The clinical utility of each model was assessed using decision curve analysis and compared with a strategy of performing SLNB on all patients. Results Among 10 089 included patients, the median (IQR) age was 64.0 (52.0-73.0) years, and 5340 (52.9%) were male. The median Breslow thickness was 1.8 mm, and 1802 patients (17.9%) had a positive SLN. Both models were well calibrated across the full range of predicted probabilities and had similar external area under the receiver operating characteristic curves (AUC; MSKCC: 70.8%; 95% CI, 69.5-72.1 and limited MIA: 69.7%; 95% CI, 68.4-71.1). At a risk threshold of 5%, decision curve analysis indicated no added net benefit for either model compared to performing SLNB for all patients. At risk thresholds of 10% or higher, both models added net benefit compared to SLNB for all patients. The greatest benefit was observed in patients with T2 melanomas using a threshold of 10%; in that setting, the use of the nomograms led to a net reduction of 8 avoidable SLNBs per 100 patients for the MSKCC nomogram and 7 per 100 patients for the limited MIA nomogram compared to a strategy of SLNB for all. Conclusions and Relevance This study confirmed the statistical performance of both the MSKCC and limited MIA models in a large, nationally representative data set. However, decision curve analysis demonstrated that using the models only improved selection for SLNB compared to biopsy in all patients when a risk threshold of at least 7% was used, with the greatest benefit seen for T2 melanomas at a threshold of 10%. Care should be taken when using these nomograms to guide selection for SLNB at the lowest thresholds.
Collapse
Affiliation(s)
- Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rasmus Mikiver
- Regional Cancer Center Southeast Sweden and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Serigne N. Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander C. J. van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel G. Coit
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christian Ingvar
- Department of Clinical Sciences, Surgery, Lund University, Lund, Sweden
| | - Karolin Isaksson
- Department of Clinical Sciences, Surgery, Lund University, Lund, Sweden
- Department of Surgery, Kristianstad Hospital, Kristianstad, Sweden
| | - Richard A. Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - John F. Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander H. R. Varey
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Plastic Surgery, Westmead Hospital, Sydney, New South Wales, Australia
| | - Sandra L. Wong
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Johan Lyth
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Edmund K. Bartlett
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
155
|
Hazard M, Duval F, Dutriaux C, Beylot-Barry M, Pham-Ledard A, Quereux G, Amini-Adle M, Heidelberger V, Aubin F, Saint-Jean M, Nardin C, Abed S, Leccia MT, Mansard S, Prey S, Khammari A, Dréno B, Gérard E. Neurological toxicities of targeted therapies in melanoma: a multicenter national observational study of the French Group of Skin Cancers (Groupe de Cancérologie Cutanée, GCC). J Neurol 2024; 271:1473-1482. [PMID: 38052771 DOI: 10.1007/s00415-023-12105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Affiliation(s)
- Madeleine Hazard
- Department of Dermatology, Hospital Dupuytren 2, UHC of Limoges, Limoges, France
| | - Fanny Duval
- Department of Medical Neurology, Hospital Pellegrin, UHC of Bordeaux, Bordeaux, France
| | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Marie Beylot-Barry
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Anne Pham-Ledard
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Gaelle Quereux
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | | | | | - François Aubin
- Department of Dermatology, UHC of Besançon, Besançon, France
| | - Mélanie Saint-Jean
- Department of Oncology, Cancer Comprehensive Center, Saint-Herblain, France
| | - Charlée Nardin
- Department of Dermatology, UHC of Besançon, Besançon, France
| | - Safia Abed
- Department of Dermatology, Military Training Hospital of Sainte Anne, Toulon, France
| | | | - Sandrine Mansard
- Department of Dermatology, Estaing Hospital, Clermont-Ferrand, France
| | - Sorilla Prey
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Amir Khammari
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | - Brigitte Dréno
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | - Emilie Gérard
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France.
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France.
| |
Collapse
|
156
|
Maher NG, Vergara IA, Long GV, Scolyer RA. Prognostic and predictive biomarkers in melanoma. Pathology 2024; 56:259-273. [PMID: 38245478 DOI: 10.1016/j.pathol.2023.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/20/2023] [Indexed: 01/22/2024]
Abstract
Biomarkers help to inform the clinical management of patients with melanoma. For patients with clinically localised primary melanoma, biomarkers can help to predict post-surgical outcome (including via the use of risk prediction tools), better select patients for sentinel lymph node biopsy, and tailor catch-all follow-up protocols to the individual. Systemic drug treatments, including immune checkpoint inhibitor (ICI) therapies and BRAF-targeted therapies, have radically improved the prognosis of metastatic (stage III and IV) cutaneous melanoma patients, and also shown benefit in the earlier setting of stage IIB/C primary melanoma. Unfortunately, a response is far from guaranteed. Here, we review clinically relevant, established, and emerging, prognostic, and predictive pathological biomarkers that refine clinical decision-making in primary and metastatic melanoma patients. Gene expression profile assays and nomograms are emerging tools for prognostication and sentinel lymph node risk prediction in primary melanoma patients. Biomarkers incorporated into clinical practice guidelines include BRAF V600 mutations for the use of targeted therapies in metastatic cutaneous melanoma, and the HLA-A∗02:01 allele for the use of a bispecific fusion protein in metastatic uveal melanoma. Several predictive biomarkers have been proposed for ICI therapies but have not been incorporated into Australian clinical practice guidelines. Further research, validation, and assessment of clinical utility is required before more prognostic and predictive biomarkers are fluidly integrated into routine care.
Collapse
Affiliation(s)
- Nigel G Maher
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
157
|
Tan SX, Chong S, Rowe C, Galbraith J, Dight J, Zhou C, Malt M, Smithers BM, Khosrotehrani K. Lymphatic expression of the proliferation marker Ki67 is linked to sentinel node positivity, recurrence and mortality in primary cutaneous melanoma. Exp Dermatol 2024; 33:e15041. [PMID: 38433382 DOI: 10.1111/exd.15041] [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: 08/03/2023] [Revised: 01/12/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
Lymphangiogenesis is a precursor to lymphovascular invasion, and may therefore signal a higher risk of metastasis and mortality in primary cutaneous melanoma. This retrospective longitudinal study aimed to evaluate whether emergent lymphangiogenesis, as measured through co-expression of endothelial proteins with the proliferation marker Ki67, was associated with poorer prognosis in a cohort of patients with single primary cutaneous melanoma. We screened all patients with a single locally invasive primary cutaneous melanoma who received sentinel lymph node biopsy at a tertiary dermatology centre in Brisbane, Australia between 1994 and 2007. Primary melanoma sections were stained via Opal multiplex immunofluorescence, and categorized according to the presence of Ki67 within either CD31+ or D2-40+ endothelial cells. Multivariate Cox regression modelling was used to evaluate associations between endothelial Ki67 positivity and clinical outcomes, with adjustment for age, sex, Breslow depth, ulceration, and anatomical location. Overall, 264 patients were available for analysis, with a median follow-up duration of 7.1 years. The presence of D2-40+ /Ki67+ co-expression was associated with greater melanoma-specific mortality (adjusted hazard ratio [HR]: 2.03; 95% confidence interval [CI]: 1.33-3.10; p = 0.001) and recurrence (adjusted HR: 1.70; 95% CI: 1.33-3.10; p = 0.001) relative to absence. CD31+ /Ki67+ co-expression was not prognostic in this cohort. Lymphatic proliferation, as measured through D2-40+ /Ki67+ co-expression, predicted greater melanoma-specific mortality and recurrence in this cohort of primary cutaneous melanoma.
Collapse
Affiliation(s)
- Samuel X Tan
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Sharene Chong
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Casey Rowe
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Jack Galbraith
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - James Dight
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Chenhao Zhou
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Maryrose Malt
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bernard Mark Smithers
- Queensland Melanoma Project, Princess Alexandra Hospital, University of Queensland, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
158
|
Smithy JW, Chapman PB. A General Approach to Patients Presenting With Locally Advanced or Distant Metastatic Disease. Cancer J 2024; 30:48-53. [PMID: 38527257 DOI: 10.1097/ppo.0000000000000704] [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: 03/27/2024]
Abstract
ABSTRACT The widespread adoption of immune checkpoint inhibitors and small molecule inhibitors of the MAP kinase pathway has transformed the management of locally advanced and metastatic melanoma. Here, we provide a broad overview on the use of these agents in the first-line setting, incorporating a review of the clinical literature as well as the practice patterns of our respective melanoma groups. Throughout, we highlight areas of uncertainty that provide opportunities for future clinical investigation and additional improvement in outcomes for patients with melanoma.
Collapse
Affiliation(s)
- James W Smithy
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | |
Collapse
|
159
|
Fortuna A, Amaral T. Multidisciplinary approach and treatment of acral and mucosal melanoma. Front Oncol 2024; 14:1340408. [PMID: 38469235 PMCID: PMC10926023 DOI: 10.3389/fonc.2024.1340408] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/13/2024] Open
Abstract
Acral and mucosal melanoma are uncommon variants of melanoma. Acral melanoma has an age-adjusted incidence of approximately 1.8 cases per million individuals per year, accounting for about 2% to 3% of all melanoma cases. On the other hand, mucosal melanoma, with an incidence of 2.2 cases per million per year, makes up around 1.3% of all melanoma cases. These melanomas, in addition to being biologically and clinically distinct from cutaneous melanoma, share certain clinical and pathologic characteristics. These include a more aggressive nature and a less favorable prognosis. Furthermore, they exhibit a different mutational pattern, with KIT mutations being more prevalent in acral and mucosal melanomas. This divergence in mutational patterns may partially account for the relatively poorer prognosis, particularly to immune checkpoint inhibitors. This review explores various aspects of acral and mucosal melanoma, including their clinical presentation, pathologic features, mutational profiles, current therapeutic approaches, outcomes associated with systemic therapy, and potential strategies to address resistance to existing treatments.
Collapse
Affiliation(s)
- Ana Fortuna
- Oncology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence Image-Guided and Functionally Instructed Tumor Therapies (iFIT) (EXC 2180), Tübingen, Germany
| |
Collapse
|
160
|
Orme SE, Moncrieff MD. A Review of Contemporary Guidelines and Evidence for Wide Local Excision in Primary Cutaneous Melanoma Management. Cancers (Basel) 2024; 16:895. [PMID: 38473257 DOI: 10.3390/cancers16050895] [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: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Surgical wide local excision (WLE) remains the current standard of care for primary cutaneous melanoma. WLE is an elective procedure that aims to achieve locoregional disease control with minimal functional and cosmetic impairment. Despite several prospective randomised trials, the optimal extent of excision margin remains controversial, and this is reflected in the persistent lack of consensus in guidelines globally. Furthermore, there is now the added difficulty of interpreting existing trial data in the context of the evolving role of surgery in the management of melanoma, with our increased understanding of clinicopathologic and genomic prognostic markers leading to the often routine use of sentinel node biopsy (SNB) as a staging procedure, in addition to the development of adjuvant systemic therapies for high-risk disease. An ongoing trial, MelMarT-II, has been designed with the aim of achieving a definitive answer to guide this fundamental surgical decision.
Collapse
Affiliation(s)
- Sophie E Orme
- Norfolk & Norwich University Hospitals NHS Foundation Trust, Norwich NR4 7UY, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Marc D Moncrieff
- Norfolk & Norwich University Hospitals NHS Foundation Trust, Norwich NR4 7UY, UK
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
161
|
Safrygina E, Applebee C, McIntyre A, Padget J, Larijani B. Spatial functional mapping of hypoxia inducible factor heterodimerisation and immune checkpoint regulators in clear cell renal cell carcinoma. BJC REPORTS 2024; 2:10. [PMID: 39516578 PMCID: PMC11524007 DOI: 10.1038/s44276-023-00033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 11/16/2024]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a highly malignant subtype of kidney cancer. Ninety percent of ccRCC have inactivating mutations of VHL that stabilise transcription factors, HIF1α and HIF2α, only stabilised in hypoxia. The varied response to HIF2 inhibition, in the preclinical and clinical settings, suggests that assessment of HIF2α activation state, not just expression levels is required as a biomarker of sensitivity to enable optimal clinical use. METHODS Two-site amplified time-resolved Förster Resonance Energy Transfer (aiFRET), with FRET-Efficiency, E f , as its read out, provides functional proteomics quantification, a precise step forward from protein expression as a tool for patient stratification. To enhance the clinical accessibility of E f , we have devised a new computational approach, Functional Oncology map (FuncOmap). RESULTS FuncOmap directly maps functional states of oncoproteins and allows functional states quantification at an enhanced spatial resolution. The innovative contributions in FuncOmap are the means to co-analyse and map expressional and functional state images and the enhancement of spatial resolution to facilitate clinical application. We show the spatial interactive states HIF2α and HIF1β in ccRCC patient samples. CONCLUSION FuncOmap can be used to quantify heterogeneity in patient response and improve accurate patient stratification, thus enhancing the power of precision.
Collapse
Affiliation(s)
- Elena Safrygina
- Cell Biophysics Laboratory, Centre for Therapeutic Innovation, Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
- ART-AI, Department of Computational Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Christopher Applebee
- Cell Biophysics Laboratory, Centre for Therapeutic Innovation, Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Alan McIntyre
- Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, Science Road, University of Nottingham, NG7 2RD, Nottingham, UK
| | - Julian Padget
- ART-AI, Department of Computational Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Banafshé Larijani
- Cell Biophysics Laboratory, Centre for Therapeutic Innovation, Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
162
|
Dugan MM, Perez MC, Karapetyan L, Zager JS. Management of acral lentiginous melanoma: current updates and future directions. Front Oncol 2024; 14:1323933. [PMID: 38390259 PMCID: PMC10882087 DOI: 10.3389/fonc.2024.1323933] [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: 10/18/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Acral lentiginous melanoma is a rare subtype of melanoma generally associated with poor outcomes, even when diagnosed at an early stage. The tumor genetic profile remains poorly understood, but it is known to have a suppressed immune environment compared to that of non-acral cutaneous melanomas, which limits therapy options. There is significant attention on the development of novel therapeutic approaches, although studies are limited due to disease rarity. For local disease, wide local excision remains the standard of care. Due to frequent under-staging on preoperative biopsy, wider margins and routine sentinel lymph node biopsy may be considered if morbidity would not be increased. For advanced disease, anti-PD1 monotherapy or combination therapy with anti-PD1 and anti-CTLA4 agents have been used as first-line treatment modalities. Anti-PD1 and anti-CTLA4 combination therapies have been shown to be particularly beneficial for patients with BRAF-mutant acral lentiginous melanoma. Other systemic combination regimens and targeted therapy options may be considered, although large studies with consistent results are lacking. Regional and intralesional therapies have shown promise for cutaneous melanomas, but studies generally have not reported results for specific histologic subtypes, especially for acral melanoma. Overall, the unique histologic and genetic characteristics of acral lentiginous melanoma make therapy options significantly more challenging. Furthermore, studies are limited, and data reporting has been inconsistent. However, more prospective studies are emerging, and alternative therapy pathways specific to acral lentiginous melanoma are being investigated. As further evidence is discovered, reliable treatment guidelines may be developed.
Collapse
Affiliation(s)
- Michelle M Dugan
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Matthew C Perez
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
163
|
Jackson-Carroll N, Johnson C, Tawbi H, Wang XS, Whisenant M. The Symptom Experience of Patients with Advanced Melanoma Undergoing Immune Checkpoint Inhibitor (ICI) Therapy. Semin Oncol Nurs 2024; 40:151574. [PMID: 38220519 DOI: 10.1016/j.soncn.2023.151574] [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: 09/28/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVES The advent of immune checkpoint inhibitor (ICI) therapy has vastly improved outcomes for patients with advanced melanoma. However, the symptom burden and intensity with their impact on quality-of-life (HRQoL) and functionality are heterogeneous and unpredictable. We used descriptive exploratory content analysis from interviews to capture the patient experience after they had completed quantitative data collection of their symptom burden and interference with the use of two patient-reported outcome (PRO) instruments. DATA SOURCES Participants from a single center with advanced melanoma (n = 19) who are undergoing ICI therapy completed the Modified MD Anderson Symptom Inventory and Functional Assessment of Cancer Therapy-Melanoma and recorded semistructured interviews. Interpretive description informed the inductive and iterative analysis approach. CONCLUSION Participants had a heterogenous experience of ICI and melanoma-related symptoms: distress (84%), fatigue (68%), rash or skin changes (53%), pain (30%), diarrhea (30%), itching (26%), and shortness of breath (21%), with varying interference within HRQoL domains, mood (47%), relations with other people (26%), and activity (21%). Some noted a lack of physical interference (79%). Uncertainty was a pervasive theme in the interviews (68%) despite the majority having positive thoughts about ICI therapy (58%) and expectations of the success of therapy (53%). The physical and emotional burden of a melanoma diagnosis, undergoing therapy, and the uncertainty of the outcomes are pervasive for patients. IMPLICATIONS FOR NURSING PRACTICE Communication surrounding the diagnosis, prognosis, treatment options, and outcomes need to be clear and acknowledge there are unknowns. Nurses may benefit from using a validated PRO instrument to help document and understand the patient's symptom experience while undergoing ICI therapy.
Collapse
Affiliation(s)
- Natalie Jackson-Carroll
- Cizik School of Nursing, The University of Texas Health Science Center at Houston; Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Constance Johnson
- Cizik School of Nursing, The University of Texas Health Science Center at Houston, Houston, TX
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xin Shelley Wang
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Meagan Whisenant
- Department of Behavioral Science, University of Texas MD Anderson Cancer Center; Cizik School of Nursing, The University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
164
|
Wan G, Leung BW, DeSimone MS, Nguyen N, Rajeh A, Collier MR, Rashdan H, Roster K, Zhou X, Moseley CB, Nirmal AJ, Pelletier RJ, Maliga Z, Marko-Varga G, Németh IB, Tsao H, Asgari MM, Gusev A, Stagner AM, Lian CG, Hurlbert MS, Liu F, Yu KH, Sorger PK, Semenov YR. Development and validation of time-to-event models to predict metastatic recurrence of localized cutaneous melanoma. J Am Acad Dermatol 2024; 90:288-298. [PMID: 37797836 PMCID: PMC10843255 DOI: 10.1016/j.jaad.2023.08.105] [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: 01/12/2023] [Revised: 08/17/2023] [Accepted: 08/28/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND The recent expansion of immunotherapy for stage IIB/IIC melanoma highlights a growing clinical need to identify patients at high risk of metastatic recurrence and, therefore, most likely to benefit from this therapeutic modality. OBJECTIVE To develop time-to-event risk prediction models for melanoma metastatic recurrence. METHODS Patients diagnosed with stage I/II primary cutaneous melanoma between 2000 and 2020 at Mass General Brigham and Dana-Farber Cancer Institute were included. Melanoma recurrence date and type were determined by chart review. Thirty clinicopathologic factors were extracted from electronic health records. Three types of time-to-event machine-learning models were evaluated internally and externally in the distant versus locoregional/nonrecurrence prediction. RESULTS This study included 954 melanomas (155 distant, 163 locoregional, and 636 1:2 matched nonrecurrences). Distant recurrences were associated with worse survival compared to locoregional/nonrecurrences (HR: 6.21, P < .001) and to locoregional recurrences only (HR: 5.79, P < .001). The Gradient Boosting Survival model achieved the best performance (concordance index: 0.816; time-dependent AUC: 0.842; Brier score: 0.103) in the external validation. LIMITATIONS Retrospective nature and cohort from one geography. CONCLUSIONS These results suggest that time-to-event machine-learning models can reliably predict the metastatic recurrence from localized melanoma and help identify high-risk patients who are most likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Guihong Wan
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Systems Biology, Harvard Medical School, Boston, Massachusetts; Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Bonnie W Leung
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mia S DeSimone
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nga Nguyen
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ahmad Rajeh
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael R Collier
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hannah Rashdan
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Katie Roster
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xu Zhou
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; School of Systems and Enterprises, Stevens Institute of Technology, Hoboken, New Jersey
| | - Cameron B Moseley
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ajit J Nirmal
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | - Roxanne J Pelletier
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | - Zoltan Maliga
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | | | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Hensin Tsao
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maryam M Asgari
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Population Medicine, Harvard Pilgrim Healthcare, Boston, Massachusetts
| | - Alexander Gusev
- Department of Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anna M Stagner
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christine G Lian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marc S Hurlbert
- Melanoma Research Alliance, Washington, District of Columbia
| | - Feng Liu
- School of Systems and Enterprises, Stevens Institute of Technology, Hoboken, New Jersey
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter K Sorger
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | - Yevgeniy R Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Systems Biology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
165
|
Chen X, Chen LJ, Peng XF, Deng L, Wang Y, Li JJ, Guo DL, Niu XH. Anti-PD-1/PD-L1 therapy for colorectal cancer: Clinical implications and future considerations. Transl Oncol 2024; 40:101851. [PMID: 38042137 PMCID: PMC10701436 DOI: 10.1016/j.tranon.2023.101851] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer in the world. The PD-1/PD-L1 pathway plays a crucial role in modulating immune response to cancer, and PD-L1 expression has been observed in tumor and immune cells within the tumor microenvironment of CRC. Thus, immunotherapy drugs, specifically checkpoint inhibitors, have been developed to target the PD-1/PD-L1 signaling pathway, thereby inhibiting the interaction between PD-1 and PD-L1 and restoring T-cell function in cancer cells. However, the emergence of resistance mechanisms can reduce the efficacy of these treatments. To counter this, monoclonal antibodies (mAbs) have been used to improve the efficacy of CRC treatments. mAbs such as nivolumab and pembrolizumab are currently approved for CRC treatment. These antibodies impede immune checkpoint receptors, including PD-1/PD-L1, and their combination therapy shows promise in the treatment of advanced CRC. This review presents a concise overview of the use of the PD-1/PD-L1 blockade as a therapeutic strategy for CRC using monoclonal antibodies and combination therapies. Additionally, this article outlines the function of PD-1/PD-L1 as an immune response suppressor in the CRC microenvironment as well as the potential advantages of administering inflammatory agents for CRC treatment. Finally, this review analyzes the outcomes of clinical trials to examine the challenges of anti-PD-1/PD-L1 therapeutic resistance.
Collapse
Affiliation(s)
- Xiang Chen
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Ling-Juan Chen
- Department of Clinical Laboratory, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Xiao-Fei Peng
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Ling Deng
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Yan Wang
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Jiu-Jiang Li
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Dong-Li Guo
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China
| | - Xiao-Hua Niu
- Department of General Surgery, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong Province 511518, China.
| |
Collapse
|
166
|
Wilson AK, Lourdault K, Ostad T, Stern S, Essner R. Is therapeutic lymph node dissection of value for lymph node recurrence in melanoma? Am J Surg 2024; 228:258-263. [PMID: 37923660 DOI: 10.1016/j.amjsurg.2023.10.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Therapeutic lymphadenectomy (TLND) is still performed in most melanoma patients to treat nodal recurrences after initial negative lymph node biopsy (-SLNB), despite the lack of evidence for survival benefit. We sought to compare melanoma-specific survival (MSS) and distant metastasis-free survival (DMFS) of patients who underwent TLND versus no TLND using our institutional and MSTL-1 databases. METHODS We identified 146 patients with nodal recurrence following -SLNB: 132 underwent TLND and 14 did not. DMFS and MSS were evaluated for the cohorts followed by a matched-pair analysis between the cohorts. RESULTS No difference was observed in five-year DMFS (p = 0.454) and five-year MSS (p = 0.945) between the two groups. The matched-pair analysis showed similar results (p = 0.329 and p = 0.363 for DMSF and MSS, respectively). CONCLUSIONS From this limited retrospective study, TLND for nodal recurrence after a -SLNB does not appear to improve DMFS or MSS in melanoma patients compared to no TLND.
Collapse
Affiliation(s)
- Ana K Wilson
- Saint John's Cancer Institute at Providence Saint John's Health Center, Department of Surgical Oncology, Santa Monica, CA, United States
| | - Kristel Lourdault
- Saint John's Cancer Institute at Providence Saint John's Health Center, Melanoma and CutaneousOncology Laboratory, Santa Monica, CA, United States
| | - Tara Ostad
- Saint John's Cancer Institute at Providence Saint John's Health Center, Melanoma and CutaneousOncology Laboratory, Santa Monica, CA, United States
| | - Stacey Stern
- Saint John's Cancer Institute at Providence Saint John's Health Center, Data Management/Biostatistics, Santa Monica, CA, United States
| | - Richard Essner
- Saint John's Cancer Institute at Providence Saint John's Health Center, Melanoma and CutaneousOncology Laboratory, Santa Monica, CA, United States.
| |
Collapse
|
167
|
Sun J, Karasaki KM, Farma JM. The Use of Gene Expression Profiling and Biomarkers in Melanoma Diagnosis and Predicting Recurrence: Implications for Surveillance and Treatment. Cancers (Basel) 2024; 16:583. [PMID: 38339333 PMCID: PMC10854922 DOI: 10.3390/cancers16030583] [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: 12/26/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Cutaneous melanoma is becoming more prevalent in the United States and has the highest mortality among cutaneous malignancies. The majority of melanomas are diagnosed at an early stage and, as such, survival is generally favorable. However, there remains prognostic uncertainty among subsets of early- and intermediate-stage melanoma patients, some of whom go on to develop advanced disease while others remain disease-free. Melanoma gene expression profiling (GEP) has evolved with the notion to help bridge this gap and identify higher- or lower-risk patients to better tailor treatment and surveillance protocols. These tests seek to prognosticate melanomas independently of established AJCC 8 cancer staging and clinicopathologic features (sex, age, primary tumor location, thickness, ulceration, mitotic rate, lymphovascular invasion, microsatellites, and/or SLNB status). While there is a significant opportunity to improve the accuracy of melanoma prognostication and diagnosis, it is equally important to understand the current landscape of molecular profiling for melanoma treatment. Society guidelines currently do not recommend molecular testing outside of clinical trials for melanoma clinical decision making, citing insufficient high-quality evidence guiding indications for the testing and interpretation of results. The goal of this chapter is to review the available literature for GEP testing for melanoma diagnosis and prognostication and understand their place in current treatment paradigms.
Collapse
Affiliation(s)
- James Sun
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19002, USA;
| | | | - Jeffrey M. Farma
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19002, USA;
| |
Collapse
|
168
|
Su J, Fu Y, Cui Z, Abidin Z, Yuan J, Zhang X, Li R, Zhao C. Relatlimab: a novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front Pharmacol 2024; 14:1349081. [PMID: 38269271 PMCID: PMC10806167 DOI: 10.3389/fphar.2023.1349081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Relatlimab is a type of human immunoglobulin G4 monoclonal blocking antibody. It is the world's first Lymphocyte-Activation Gene-3 (LAG-3) inhibitor and the third immune checkpoint inhibitor with clinical application, following PD-1 and CTLA-4. Relatlimab can bind to the LAG-3 receptor which blocks the interaction between LAG-3 and its ligand to reduce LAG-3 pathway-mediated immunosuppression and promote T-cell proliferation, inducing tumor cell death. On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab developed by Bristol Myers Squibb with nivolumab, under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adult and pediatric patients aged 12 and older. This study comprehensively describes the mechanism of action and clinical trials of relatlimab and a brief overview of immune checkpoint drugs currently used for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Yiting Fu
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zitong Cui
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zain Abidin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jingsong Yuan
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Xinmiao Zhang
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Runmin Li
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
169
|
Monti M, Ferrari G, Grosso V, Missale F, Bugatti M, Cancila V, Zini S, Segala A, La Via L, Consoli F, Orlandi M, Valerio A, Tripodo C, Rossato M, Vermi W. Impaired activation of plasmacytoid dendritic cells via toll-like receptor 7/9 and STING is mediated by melanoma-derived immunosuppressive cytokines and metabolic drift. Front Immunol 2024; 14:1227648. [PMID: 38239354 PMCID: PMC10795195 DOI: 10.3389/fimmu.2023.1227648] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Plasmacytoid dendritic cells (pDCs) infiltrate a large set of human cancers. Interferon alpha (IFN-α) produced by pDCs induces growth arrest and apoptosis in tumor cells and modulates innate and adaptive immune cells involved in anti-cancer immunity. Moreover, effector molecules exert tumor cell killing. However, the activation state and clinical relevance of pDCs infiltration in cancer is still largely controversial. In Primary Cutaneous Melanoma (PCM), pDCs density decreases over disease progression and collapses in metastatic melanoma (MM). Moreover, the residual circulating pDC compartment is defective in IFN-α production. Methods The activation of tumor-associated pDCs was evaluated by in silico and microscopic analysis. The expression of human myxovirus resistant protein 1 (MxA), as surrogate of IFN-α production, and proximity ligation assay (PLA) to test dsDNA-cGAS activation were performed on human melanoma biopsies. Moreover, IFN-α and CXCL10 production by in vitro stimulated (i.e. with R848, CpG-A, ADU-S100) pDCs exposed to melanoma cell lines supernatants (SN-mel) was tested by intracellular flow cytometry and ELISA. We also performed a bulk RNA-sequencing on SN-mel-exposed pDCs, resting or stimulated with R848. Glycolytic rate assay was performed on SN-mel-exposed pDCs using the Seahorse XFe24 Extracellular Flux Analyzer. Results Based on a set of microscopic, functional and in silico analyses, we demonstrated that the melanoma milieu directly impairs IFN-α and CXCL10 production by pDCs via TLR-7/9 and cGAS-STING signaling pathways. Melanoma-derived immunosuppressive cytokines and a metabolic drift represent relevant mechanisms enforcing pDC-mediated melanoma escape. Discussion These findings propose a new window of intervention for novel immunotherapy approaches to amplify the antitumor innate immune response in cutaneous melanoma (CM).
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgia Ferrari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valentina Grosso
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Nederlands Kanker Instituut, Amsterdam, Netherlands
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Stefania Zini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Agnese Segala
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Consoli
- Oncology Unit, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili di Brescia, Brescia, Italy
| | - Matteo Orlandi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
170
|
Basilicata M, Terrano V, D’Aurelio A, Bruno G, Troiani T, Bollero P, Napolitano S. Oral Adverse Events Associated with BRAF and MEK Inhibitors in Melanoma Treatment: A Narrative Literature Review. Healthcare (Basel) 2024; 12:105. [PMID: 38201012 PMCID: PMC10778825 DOI: 10.3390/healthcare12010105] [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: 11/07/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Melanoma cancer represents the most lethal type of skin cancer originating from the malignant transformation of melanocyte cells. Almost 50% of melanomas show the activation of BRAF mutations. The identification and characterization of BRAF mutations led to the development of specific drugs that radically changed the therapeutic approach to melanoma. METHODS We conducted a narrative review of the literature according to a written protocol before conducting the study. This article is based on previously conducted studies. We identified articles by searching electronic databases (Medline, Google Scholar and PubMed). We used a combination of "melanoma", "Braf-Mek inhibitors", " targeted therapy" and "oral side effects". RESULTS Eighteen studies were reported in this article showing the relationship between the use of targeted therapy in melanoma cancer and the development of oral side effects, such as mucositis, hyperkeratosis and cellular proliferation. CONCLUSION Targeted therapy plays an important role in the treatment of melanoma cancer, showing a notable increase in response rate, prolonged progression-free survival and overall survival in BRAF-mutated melanoma patients. Oral side effects represent a common finding over the course of treatment. However, these adverse effects can be easily managed in a multidisciplinary approach involving collaboration between medical oncologists and dental doctors.
Collapse
Affiliation(s)
- Michele Basilicata
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
- UniCamillus-Saint Camillus, International University of Health Sciences, 00131 Rome, Italy
| | - Vincenzo Terrano
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| | - Alessandro D’Aurelio
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
| | - Giovanni Bruno
- Department of Neuroscience, University of Padua, 35121 Padova, Italy
- Department of Industrial Engineering, University of Tor Vergata, 00133 Rome, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| | - Patrizio Bollero
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| |
Collapse
|
171
|
Moreno-Ramírez D, Fernández-Orland A, Ferrándiz L. Immunotherapy and Targeted Therapy in Patients With Advanced Melanoma and the V600 BRAF Mutation: Which One First? ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:48-55. [PMID: 37321549 DOI: 10.1016/j.ad.2023.05.023] [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: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.
Collapse
Affiliation(s)
- D Moreno-Ramírez
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España.
| | - A Fernández-Orland
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - L Ferrándiz
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España
| |
Collapse
|
172
|
Huynh J, Leiter U, Garbe C, Shiderova G, Walter V, Eigentler T, Scheu A, Häfner HM, Schnabl SM. Sentinel lymph node biopsy for lentigo maligna melanoma under local anaesthesia. J Eur Acad Dermatol Venereol 2024; 38:84-92. [PMID: 37611257 DOI: 10.1111/jdv.19456] [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: 02/02/2023] [Accepted: 08/03/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Lentigo maligna melanoma is mainly localized in the head and neck region in elderly patients. Due to its slow horizontal growth, it has a good prognosis compared to other melanoma subtypes, but specific data are rare. OBJECTIVES The aim of this study was to investigate sentinel lymph node biopsy in lentigo maligna melanoma under local anaesthesia and to discuss the benefit. METHODS Investigation of patients with lentigo maligna melanoma and tumour thickness ≥1 mm treated at the Department of Dermatology, University Medical Centre Tuebingen, between January 2008 and October 2019. RESULTS In total, 204 patients (126 SLNB, 78 non-SLNB) with a median age of 75.7 years (SLNB: 73.3 years, non-SLNB: 79.7 years) could be included. Sixteen of 126 (12.7%) sentinel lymph nodes were positive. Five-year overall survival was 87.9% (88.5% SLNB; 87.4% non-SLNB) and 5-year distant metastasis-free survival was 85.8% (85.4% SLNB; 86.7% non-SLNB). There was no significant difference for distant metastasis-free survival (p = 0.861) and overall survival (p = 0.247) between patients with and without sentinel lymph node biopsy. CONCLUSIONS Sentinel lymph node biopsy in lentigo maligna melanoma under local anaesthesia is a safe and simple method, even in very old patients. However, LMM has a very good 5-year overall survival. In high-risk patients with high tumour thickness and/or ulceration, adjuvant immunotherapy can now be offered without the need to perform this procedure.
Collapse
Affiliation(s)
- Julia Huynh
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Leiter
- Department of Dermatology, University of Tuebingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, University of Tuebingen, Tübingen, Germany
| | - Galina Shiderova
- Department of Dermatology, University of Tuebingen, Tübingen, Germany
| | - Vincent Walter
- Department of Dermatology, University of Tuebingen, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Scheu
- Department of Dermatology, University of Tuebingen, Tübingen, Germany
| | | | | |
Collapse
|
173
|
Moreno-Ramírez D, Fernández-Orland A, Ferrándiz L. [Translated article] Immunotherapy and Targeted Therapy in Patients With Advanced Melanoma and the V600 BRAF Mutation: Which One First? ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T48-T55. [PMID: 37923078 DOI: 10.1016/j.ad.2023.10.036] [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: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 11/07/2023] Open
Abstract
Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.
Collapse
Affiliation(s)
- D Moreno-Ramírez
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain.
| | - A Fernández-Orland
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - L Ferrándiz
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain
| |
Collapse
|
174
|
Du Y, Qi Z, Liang X, Dai J, Wei X, Bai X, Mao L, Chi Z, Cui C, Lian B, Tang B, Wang X, Chen Y, Guo J, Si L. Pembrolizumab Versus High-Dose Interferon-α2b as Adjuvant Therapy for Pediatric Melanoma: A Retrospective Study. Dermatol Pract Concept 2024; 14:dpc.1401a26. [PMID: 38364395 PMCID: PMC10868960 DOI: 10.5826/dpc.1401a26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 02/18/2024] Open
Abstract
INTRODUCTION Pembrolizumab is well-tolerated in pediatric patients with advanced tumors, consistent with results in adults. However, information on the safety and efficacy of adjuvant pembrolizumab in children and adolescents with melanoma is lacking. OBJECTIVES To compare pembrolizumab versus high-dose interferon-α2b (HDI) as adjuvant therapy in pediatric patients with melanoma. METHODS We performed a retrospective study of pediatric patients diagnosed with melanoma between January 2008 and April 2022. Relapse-free survival (RFS) and the 1-year RFS rate were compared between patients receiving adjuvant pembrolizumab or HDI. RESULTS Seventy-five pediatric patients with melanoma were screened from a database of 6,013 patients. Twenty-four patients were finally enrolled, of whom 9 received pembrolizumab and 15 received HDI as adjuvant therapy. By August 31, 2022, the median follow-up times were 23.6 months and 98.7 months in the pembrolizumab and HDI groups, respectively. There was no significant difference in median RFS between two groups (not reached versus 38.7 months, P = 0.11). The median overall survival was not reached in either group. The 1-year RFS rates were 88.9% and 66.7% in the pembrolizumab and HDI groups, respectively. All adverse events in the pembrolizumab group were grade 1 or 2, but grade 3-5 adverse events occurred in two (13%) patients receiving HDI. CONCLUSIONS RFS was similar in pediatric patients with melanoma receiving adjuvant pembrolizumab or HDI, but pembrolizumab was associated with a reduced risk of recurrence and a more favorable safety profile. However, due to the small sample size and differences in follow-up time, larger and prospective studies are still warranted to explore better adjuvant therapies for pediatric melanoma.
Collapse
Affiliation(s)
- Yu Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhonghui Qi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xianbin Liang
- Department of Oncology, The Third People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoting Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xue Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bin Lian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bixia Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xuan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
175
|
van Akkooi ACJ. Relatlimab, an Immune Checkpoint Inhibitor that Blocks LAG-3, the Latest Drug to be Added to the Arsenal of Systemic Therapies for Melanoma: What Does a Surgical Oncologist Need to Know? Ann Surg Oncol 2024; 31:1-3. [PMID: 37843663 DOI: 10.1245/s10434-023-14416-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Affiliation(s)
- Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
176
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
177
|
Green CE, Chacon J, Godinich BM, Hock R, Kiesewetter M, Raynor M, Marwaha K, Maharaj S, Holland N. The Heart of the Matter: Immune Checkpoint Inhibitors and Immune-Related Adverse Events on the Cardiovascular System. Cancers (Basel) 2023; 15:5707. [PMID: 38136253 PMCID: PMC10742007 DOI: 10.3390/cancers15245707] [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: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer remains a prominent global cause of mortality, second only to cardiovascular disease. The past decades have witnessed substantial advancements in anti-cancer therapies, resulting in improved outcomes. Among these advancements, immunotherapy has emerged as a promising breakthrough, leveraging the immune system to target and eliminate cancer cells. Despite the remarkable potential of immunotherapy, concerns have arisen regarding associations with adverse cardiovascular events. This review examines the complex interplay between immunotherapy and cardiovascular toxicity and provides an overview of immunotherapy mechanisms, clinical perspectives, and potential biomarkers for adverse events, while delving into the intricate immune responses and evasion mechanisms displayed by cancer cells. The focus extends to the role of immune checkpoint inhibitors in cancer therapy, including CTLA-4, PD-1, and PD-L1 targeting antibodies. This review underscores the multifaceted challenges of managing immunotherapy-related cardiovascular toxicity. Risk factors for immune-related adverse events and major adverse cardiac events are explored, encompassing pharmacological, treatment-related, autoimmune, cardiovascular, tumor-related, social, genetic, and immune-related factors. The review also advocates for enhanced medical education and risk assessment tools to identify high-risk patients for preventive measures. Baseline cardiovascular evaluations, potential prophylactic strategies, and monitoring of emerging toxicity symptoms are discussed, along with the potential of adjunct anti-inflammatory therapies.
Collapse
Affiliation(s)
- Chase E. Green
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Jessica Chacon
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Brandon M. Godinich
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Rivers Hock
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Maria Kiesewetter
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Mark Raynor
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Komal Marwaha
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Satish Maharaj
- Department of Internal Medicine, Division of Hematology/Oncology, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 4800 Alberta Ave., El Paso, TX 79905, USA
| | - Nathan Holland
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| |
Collapse
|
178
|
You X, Koop K, Weigert A. Heterogeneity of tertiary lymphoid structures in cancer. Front Immunol 2023; 14:1286850. [PMID: 38111571 PMCID: PMC10725932 DOI: 10.3389/fimmu.2023.1286850] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
The success of immunotherapy approaches, such as immune checkpoint blockade and cellular immunotherapy with genetically modified lymphocytes, has firmly embedded the immune system in the roadmap for combating cancer. Unfortunately, the majority of cancer patients do not yet benefit from these therapeutic approaches, even when the prognostic relevance of the immune response in their tumor entity has been demonstrated. Therefore, there is a justified need to explore new strategies for inducing anti-tumor immunity. The recent connection between the formation of ectopic lymphoid aggregates at tumor sites and patient prognosis, along with an effective anti-tumor response, suggests that manipulating the occurrence of these tertiary lymphoid structures (TLS) may play a critical role in activating the immune system against a growing tumor. However, mechanisms governing TLS formation and a clear understanding of their substantial heterogeneity are still lacking. Here, we briefly summarize the current state of knowledge regarding the mechanisms driving TLS development, outline the impact of TLS heterogeneity on clinical outcomes in cancer patients, and discuss appropriate systems for modeling TLS heterogeneity that may help identify new strategies for inducing protective TLS formation in cancer patients.
Collapse
Affiliation(s)
- Xin You
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
| | - Kristina Koop
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Weigert
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
179
|
Thompson JF. Harold Gillies, pioneer of modern plastic surgery, and Donald Morton, pioneer of modern surgical oncology: Master problem-solvers and surgical role models. J Plast Reconstr Aesthet Surg 2023; 87:24-32. [PMID: 37804644 DOI: 10.1016/j.bjps.2023.09.047] [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: 08/01/2023] [Accepted: 09/12/2023] [Indexed: 10/09/2023]
Abstract
Harold Gillies, plastic surgeon, and Donald Morton, surgical oncologist, were iconic pioneers in their respective fields. Both of them made their mark by identifying crucial practical problems and finding innovative ways of solving them. Gillies grappled with the challenge of restoring form and function to British military personnel injured in World War I, and he set up a dedicated facility for performing this work. He introduced many new reconstructive techniques that became the foundation of the modern specialty of plastic and reconstructive surgery, which he established and nurtured. Morton, in the United States, applied his problem-solving skills to the long-debated question of the best way to manage regional lymph nodes in patients with melanoma. He developed the innovative technique of sentinel lymph node biopsy and initiated large-scale international clinical trials to establish its validity and clinical value. This and other important contributions to the emerging field of surgical oncology earned Morton his reputation as a pioneer and leader of that specialty. The problems that confronted Gillies and Morton were completely different, but both demonstrated remarkable skills as master problem-solvers in their respective fields and made extraordinary contributions to the body of knowledge and welfare of patients. All surgeons must be problem-solvers because every patient who presents for surgical management represents a new problem (or set of problems) to be addressed. As surgeons, we would do well to consider individuals such as Gillies and Morton as role models for our own problem-solving activities in day-to-day clinical practice.
Collapse
Affiliation(s)
- J F Thompson
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Division of Surgery, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Faculty of Health and Medical Sciences, The University of Western Australia.
| |
Collapse
|
180
|
Walia A, Tuia J, Prasad V. Progression-free survival, disease-free survival and other composite end points in oncology: improved reporting is needed. Nat Rev Clin Oncol 2023; 20:885-895. [PMID: 37828154 DOI: 10.1038/s41571-023-00823-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Composite outcome measures such as progression-free survival and disease-free survival are increasingly used as surrogate end points in oncology research, frequently serving as the primary end point of pivotal trials that form the basis for FDA and EMA approvals. Such outcome measures combine two or more distinct events (for example, tumour (re)growth, new lesions and/or death) into a single, time-to-event end point. The use of a composite end point can increase the statistical power of a clinical trial and decrease the follow-up period required to demonstrate efficacy, thus lowering costs; however, these end points have a number of limitations. Composite outcomes are often vaguely defined, with definitions that vary greatly between studies, complicating comparisons of results across trials. Altering the makeup of events included in a composite outcome can alter study conclusions, including whether treatment effects are statistically significant. Moreover, the events included in a composite outcome often vary in clinical significance, reflect distinct biological pathways and/or are affected differently by treatment. Therefore, knowing the precise breakdown of the component events is essential to accurately interpret trial results and gauge the true benefit of an intervention. In oncology clinical trials, however, such information is rarely provided. In this Perspective, we emphasize this deficiency through a review of 50 studies with progression-free survival as an outcome published in five top oncology journals, discuss the advantages and challenges of using composite end points, and highlight the need for transparent reporting of the component events.
Collapse
Affiliation(s)
- Anushka Walia
- School of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Jordan Tuia
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
181
|
Taushanova MS, Milusheva YI, Manov DA, Hadjieva RR, Yordanov AD. Synchronous Occurrence of Triple-Negative Breast Cancer and Malignant Melanoma. J Med Cases 2023; 14:400-404. [PMID: 38186558 PMCID: PMC10769651 DOI: 10.14740/jmc4167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
In people with cancer, multiple primary malignant neoplasms (MPMNs) are not unusual, and they may be caused by risk factors such as genetics, viral infection, smoking, environmental factors, or treatment-related variables. The frequency of MPMNs occurring in the same or separate organ systems is between 2% and 17%. The 5-year breast cancer survivors have been found to have around 3.6% chance of acquiring another neoplasm. In this case report, we present a very rare simultaneous occurrence of two highly malignant tumors - triple-negative breast cancer and cutaneous melanoma. We performed genetic tests for determining the link between both neoplasms. The patient was treated in an adjuvant setting with chemotherapy and immunotherapy with pembrolizumab. According to epidemiological studies, for primary cutaneous melanoma following breast cancer, the standardized incidence ratio (SIR) varied from 1.03 to 4.10, while for primary breast carcinoma following cutaneous melanoma, it varied from 1.16 to 5.13. A number of risk factors have been proven to increase the risk of a second primary malignancy. This case highlights the importance of risk factor assessment and thorough primary workup of each patient. It emphasizes the need for a personalized approach when treating synchronous neoplasms.
Collapse
Affiliation(s)
| | - Yoana Ivanova Milusheva
- Department of Medical Oncology, University Hospital “Tsaritsa Yoanna-ISUL”, 1000 Sofia, Bulgaria
| | - Dimo Angelov Manov
- Department of Medical Oncology, MBAL “Nadezhda Hospital”, 1000 Sofia, Bulgaria
| | | | | |
Collapse
|
182
|
Karch JL, Davis MJ, Momtahen S, Simmons BJ. Subcutaneous Nodule With Poliosis: An Unusual Presentation of Melanoma Ex Blue Nevus. Dermatol Surg 2023; 49:1206-1207. [PMID: 37738284 DOI: 10.1097/dss.0000000000003951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Affiliation(s)
- Jamie L Karch
- Dartmouth Geisel School of Medicine, Hanover, New Hampshire
| | - Matthew J Davis
- Dartmouth Health Medical Center, Department of Dermatology, Lebanon, New Hampshire
| | - Shabnam Momtahen
- Dartmouth Health Medical Center, Department of Pathology, Lebanon, New Hampshire
| | - Brian J Simmons
- Dartmouth Health Medical Center, Department of Dermatology, Lebanon, New Hampshire
| |
Collapse
|
183
|
Alvarez J, Smith JJ. Anorectal mucosal melanoma. SEMINARS IN COLON AND RECTAL SURGERY 2023; 34:100990. [PMID: 38746826 PMCID: PMC11090490 DOI: 10.1016/j.scrs.2023.100990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Anorectal mucosal melanoma accounts for less than 1 % of all anorectal malignant tumors and a tendency for delayed diagnosis leads to advanced disease at presentation.1,2 Due to the rarity of the disease, there are limited prospective trials exploring the optimal treatment strategies. Generally, tumors are surgically excised, with a preference for conservative management with wide local excision. In the past decade, there have been advances with immunotherapy and other targeted therapies. Multiple clinical trials continue exploring neoadjuvant/adjuvant combination treatments in the setting of advanced or unresectable disease.
Collapse
Affiliation(s)
- Janet Alvarez
- Research Scholar, Memorial Sloan Kettering Cancer Center, Department of Surgery, 1275 York Avenue | SR-201, New York, NY 10065, USA
| | - J. Joshua Smith
- Associate Member, Associate Attending Surgeon, Memorial Sloan Kettering Cancer Center, Colorectal Service, Department of Surgery, 1275 York Avenue, SR-201, New York, NY 10065, USA
| |
Collapse
|
184
|
Reticker-Flynn NE, Engleman EG. Lymph nodes: at the intersection of cancer treatment and progression. Trends Cell Biol 2023; 33:1021-1034. [PMID: 37149414 PMCID: PMC10624650 DOI: 10.1016/j.tcb.2023.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
Metastasis to lymph nodes (LNs) is a common feature of disease progression in most solid organ malignancies. Consequently, LN biopsy and lymphadenectomy are common clinical practices, not only because of their diagnostic utility but also as a means of deterring further metastatic spread. LN metastases have the potential to seed additional tissues and can induce metastatic tolerance, a process by which tumor-specific immune tolerance in LNs promotes further disease progression. Nonetheless, phylogenetic studies have revealed that distant metastases are not necessarily derived from nodal metastases. Furthermore, immunotherapy efficacy is increasingly being attributed to initiation of systemic immune responses within LNs. We argue that lymphadenectomy and nodal irradiation should be approached with caution, particularly in patients receiving immunotherapy.
Collapse
Affiliation(s)
- Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
185
|
Quan Q, Guo L, Huang L, Liu Z, Guo T, Shen Y, Ding S, Liu C, Cao L. Expression and clinical significance of PD-L1 and infiltrated immune cells in the gastric adenocarcinoma microenvironment. Medicine (Baltimore) 2023; 102:e36323. [PMID: 38050283 PMCID: PMC10695517 DOI: 10.1097/md.0000000000036323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/03/2023] [Indexed: 12/06/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial negative costimulatory molecule expressed on both tumor and immune cells. It binds to programmed death-1, facilitating tumor escape. Tumor-infiltrating immune cells play a vital role in this process. However, the clinical relationship between PD-L1 expression and tumor-infiltrating immune cells remains uncertain. Immunohistochemistry (IHC) was utilized to assess PD-L1 expression and TIIC markers (CD3, CD4, CD8, CD19, CD31, CD68, CD11c, CD56, and α-smooth muscle actin) in gastric adenocarcinoma tissues from 268 patients. The aim was to explore the prognostic significance of PD-L1 and the infiltration of different immune cell types. The study analyzed overall survival and the correlations between PD-L1 expression, immune cell infiltration, and clinicopathological characteristics. Among the 268 patients, 52 (19.40%) exhibited high PD-L1 expression on tumor cells (TPD-L1), while 167 (62.31%) displayed high PD-L1 expression on immune cells (IPD-L1). Patients with high IPD-L1 expression showed improved survival compared to those with low IPD-L1 expression (P = .028). High TPD-L1 expression associated with various clinicopathological features, such as larger tumor size, poorer differentiation, deeper invasion depth, and higher tumor stage. Conversely, patients with high IPD-L1 expression exhibited shallower tumor invasion and lower mortality rates. Univariate analysis indicated that superficial tumor infiltration, absence of lymph node and distant metastasis, low tumor stage, high IPD-L1 expression, and elevated CD8 and CD19 expression were associated with a reduced risk of tumor progression. Multivariate analysis revealed that patients with high IPD-L1 and CD8 expression or high TPD-L1 and low CD31 expression experienced significantly better overall survival than patients with other combinations. The findings indicate that patients with high PD-L1 expression in immune cells have a substantially improved prognosis. Additionally, the combination of PD-L1 with CD8 or CD31 expression status can serve as an indicator of prognosis in patients with gastric adenocarcinoma.
Collapse
Affiliation(s)
- Qiuying Quan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lili Huang
- Department of Clinical Laboratory, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhiju Liu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianwei Guo
- Department of Pathology, Changshu Hospital of Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sisi Ding
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Cuiping Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lei Cao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, Suzhou, Jiangsu, China
| |
Collapse
|
186
|
Fastner S, Shen N, Hartman RI, Chu EY, Kim CC, Kirkwood JM, Grossman D. Prognostic gene expression profile testing to inform use of adjuvant therapy: A survey of melanoma experts. Cancer Med 2023; 12:22103-22108. [PMID: 38098216 PMCID: PMC10757117 DOI: 10.1002/cam4.6819] [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: 08/16/2023] [Revised: 10/16/2023] [Accepted: 12/03/2023] [Indexed: 12/31/2023] Open
Abstract
OBJECTIVES To investigate current practices and attitudes regarding use of adjuvant immunotherapy and prognostic gene expression profile (GEP) testing among melanoma medical and surgical oncologists. METHODS An anonymous RedCap-based survey was emailed to ~300 melanoma experts. RESULTS Respondents generally favored adjuvant immunotherapy over observation (73% for all Stage IIIA, 50% for Stage IIB/IIC) and cited a minimum 10-year recurrence risk of 11%-20% (48%) or 21%-30% (33%) to justify treatment, but acknowledged that risks of serious adverse events may outweigh potential benefits for some Stage IIB/IIC patients. While GEP test results did not strongly influence decision-making regarding follow-up or intervention, most were receptive to randomized trials using GEP testing to identify subsets of Stage IIB/IIC (74%) and Stage IB/IIA (54%) patients who may not or may, respectively, benefit from adjuvant therapy. CONCLUSION Although most respondents do not routinely use GEP testing, many would participate in clinical trials to determine clinical utility.
Collapse
Affiliation(s)
- Suzanne Fastner
- Huntsman Cancer InstituteUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
| | - Nathan Shen
- Huntsman Cancer InstituteUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
| | - Rebecca I. Hartman
- Department of DermatologyUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
- Department of DermatologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Department of DermatologyVA Integrated Service Network (VISN‐1)Jamaica PlainMassachusettsUSA
| | - Emily Y. Chu
- Department of DermatologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Caroline C. Kim
- Department of DermatologyTufts Medical CenterBostonMassachusettsUSA
| | - John M. Kirkwood
- Departments of DermatologyUniversity of Pittsburgh, UPMC Hillman Cancer CenterPittsburghPennsylvaniaUSA
- Departments of MedicineUniversity of Pittsburgh, UPMC Hillman Cancer CenterPittsburghPennsylvaniaUSA
| | - Douglas Grossman
- Huntsman Cancer InstituteUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
- Department of DermatologyUniversity of Utah Health Sciences CenterSalt Lake CityUtahUSA
| |
Collapse
|
187
|
Holmberg CJ, Mikiver R, Isaksson K, Ingvar C, Moncrieff M, Nielsen K, Ny L, Lyth J, Olofsson Bagge R. Prognostic Significance of Sentinel Lymph Node Status in Thick Primary Melanomas (> 4 mm). Ann Surg Oncol 2023; 30:8026-8033. [PMID: 37574516 PMCID: PMC10625939 DOI: 10.1245/s10434-023-14050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 06/12/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND The key prognostic factors for staging patients with primary cutaneous melanoma are Breslow thickness, ulceration, and sentinel lymph node (SLN) status. The multicenter selective lymphadenectomy trial (MSLT-I) verified SLN status as the most important prognostic factor for patients with intermediate-thickness melanoma (Breslow thickness, 1-4 mm). Although most international guidelines recommend SLN biopsy (SLNB) also for patients with thick (> 4 mm, pT4) melanomas, its prognostic role has been questioned. The primary aim of this study was to establish whether SLN status is prognostic in T4 melanoma tumors. METHODS Data for all patients with a diagnosis of primary invasive cutaneous melanoma of Breslow thickness greater than 1 mm in Sweden between 2007 and 2020 were retrieved from the Swedish Melanoma Registry, a large prospective population-based registry. A multivariable Cox proportional hazard model for melanoma-specific survival (MSS) was constructed based on Breslow thickness stratified for SLN status. RESULTS The study enrolled 10,491 patients, 1943 of whom had a Breslow thickness greater than 4 mm (pT4). A positive SLN was found for 34% of these pT4 patients. The 5-year MSS was 71%, and the 10-year MSS was 62%. There was a statistically significant difference in MSS between the patients with a positive SLN and those with a negative SLN (hazard ratio of 2.4 (95% confidence interval CI 1.6-3.5) for stage T4a and 2.0 (95% CI 1.6-2.5) for satage T4b. CONCLUSION Sentinel lymph node status gives important prognostic information also for patients with thick (> 4 mm) melanomas, and the authors thus recommend that clinical guidelines be updated to reflect this.
Collapse
Affiliation(s)
- Carl-Jacob Holmberg
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rasmus Mikiver
- Department of Clinical and Experimental Medicine, Regional Cancer Center Southeast Sweden, Linköping University, Linköping, Sweden
| | - Karolin Isaksson
- Department of Surgery, Kristianstad Hospital, Kristianstad, Sweden
- Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund University Cancer Centre, Lund University, Lund, Sweden
| | - Christian Ingvar
- Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marc Moncrieff
- Department of Plastic and Reconstructive Surgery, Norfolk and Norwich University Hospital, Norwich, UK
| | - Kari Nielsen
- Lund University Cancer Centre, Lund University, Lund, Sweden
- Department of Dermatology, Skåne University Hospital, Lund, Sweden
- Division of Dermatology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Lyth
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
188
|
Sheng F, Yan Y, Zeng B. Efficacy and safety of immune checkpoint inhibitors and targeted therapies in resected melanoma: a systematic review and network meta-analysis. Front Pharmacol 2023; 14:1284240. [PMID: 38026956 PMCID: PMC10661889 DOI: 10.3389/fphar.2023.1284240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Multiple immune checkpoint inhibitors (ICIs) and targeted therapies have been widely used as adjuvant treatments for high-risk resected melanoma, with unclear comparative efficacy and safety. Methods: PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov were searched from database inception until 6 June 2023. We included RCTs that assess adjuvant ICIs or targeted therapies in high-risk resected melanoma. Frequentist random-effect network meta-analyses (NMA) were performed. The primary outcome was recurrence-free survival (RFS). Results: Eleven trials including 10,712 patients and comparing 10 treatments (nivolumab [Nivo], ipilimumab 3 mg/kg [Ipi3], Ipi10, pembrolizumab [Pemb], vemurafenib [Vemu], bevacizumab [Beva], Nivo + Ipi1, Nivo + Ipi3, dabrafenib plus trametinib [Dab + Tram], and placebo/observation [Pla/Obs]) were included. NMA showed that all treatments showed RFS benefit over placebo/observation except Ipi3 (hazard ratio [HR], 0.78; 95% CI, 0.58-1.05). Combination therapy of Nivo + Ipi3 was the most effective treatment, which significantly improved RFS compared with other treatments. NMA also showed that all treatments were associated with an increased risk of grade 3-5 adverse events over placebo/observation except Nivo (HR, 1.25; 95% CI, 0.87-1.80). NMA suggested that Nivo and Pemb were the two safest treatments except for placebo/observation. Although three combination therapies ranked as the top three in terms of RFS, they did not show significant overall survival benefits compared to monotherapies including Pemb, Nivo, Ipi3, and Ipi10. Conclusion: In this NMA, adjuvant Nivo and Pemb are the preferred options in patients with resected melanoma considering the benefits and harms. Combination therapy of Nivo + Ipi3 may be a promising strategy, but more evidence from phase 3 trials is needed. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=438667, PROSPERO (CRD42023438667).
Collapse
Affiliation(s)
- Feng Sheng
- Department of Dermatology, Peking University Binhai Hospital, Tianjin, China
| | - Yulan Yan
- Hematology and Oncology, Peking University Binhai Hospital, Tianjin, China
| | - Baoqi Zeng
- Central Laboratory, Peking University Binhai Hospital, Tianjin, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| |
Collapse
|
189
|
Hoeijmakers LL, Reijers ILM, Blank CU. Biomarker-Driven Personalization of Neoadjuvant Immunotherapy in Melanoma. Cancer Discov 2023; 13:2319-2338. [PMID: 37668337 DOI: 10.1158/2159-8290.cd-23-0352] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/27/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
The introduction of immunotherapy has ushered in a new era of anticancer therapy for many cancer types including melanoma. Given the increasing development of novel compounds and combinations and the investigation in earlier disease stages, the need grows for biomarker-based treatment personalization. Stage III melanoma is one of the front-runners in the neoadjuvant immunotherapy field, facilitating quick biomarker identification by its immunogenic capacity, homogeneous patient population, and reliable efficacy readout. In this review, we discuss potential biomarkers for response prediction to neoadjuvant immunotherapy, and how the neoadjuvant melanoma platform could pave the way for biomarker identification in other tumor types. SIGNIFICANCE In accordance with the increasing rate of therapy development, the need for biomarker-driven personalized treatments grows. The current landscape of neoadjuvant treatment and biomarker development in stage III melanoma can function as a poster child for these personalized treatments in other tumors, assisting in the development of new biomarker-based neoadjuvant trials. This will contribute to personalized benefit-risk predictions to identify the most beneficial treatment for each patient.
Collapse
Affiliation(s)
- Lotte L Hoeijmakers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| | - Irene L M Reijers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Molecular Oncology and Immunology, Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands
| |
Collapse
|
190
|
Kirkwood JM, Del Vecchio M, Weber J, Hoeller C, Grob JJ, Mohr P, Loquai C, Dutriaux C, Chiarion-Sileni V, Mackiewicz J, Rutkowski P, Arenberger P, Quereux G, Meniawy TM, Ascierto PA, Menzies AM, Durani P, Lobo M, Campigotto F, Gastman B, Long GV. Adjuvant nivolumab in resected stage IIB/C melanoma: primary results from the randomized, phase 3 CheckMate 76K trial. Nat Med 2023; 29:2835-2843. [PMID: 37845511 PMCID: PMC10667090 DOI: 10.1038/s41591-023-02583-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/08/2023] [Indexed: 10/18/2023]
Abstract
Patients with resected stage IIB/C melanoma have high recurrence risk, similar to those with resected stage IIIA/B disease. The phase 3, double-blind CheckMate 76K trial assessed 790 patients with resected stage IIB/C melanoma randomized 2:1 (stratified by tumor category) to nivolumab 480 mg or placebo every 4 weeks for 12 months. The primary endpoint was investigator-assessed recurrence-free survival (RFS). Secondary endpoints included distant metastasis-free survival (DMFS) and safety. At 7.8 months of minimum follow-up, nivolumab significantly improved RFS versus placebo (hazard ratio (HR) = 0.42; 95% confidence interval (CI): 0.30-0.59; P < 0.0001), with 12-month RFS of 89.0% versus 79.4% and benefit observed across subgroups; DMFS was also improved (HR = 0.47; 95% CI: 0.30-0.72). Treatment-related grade 3/4 adverse events occurred in 10.3% (nivolumab) and 2.3% (placebo) of patients. One treatment-related death (0.2%) occurred with nivolumab. Nivolumab is an effective and generally well-tolerated adjuvant treatment in patients with resected stage IIB/C melanoma. ClinicalTrials.gov identifier: NCT04099251 .
Collapse
Affiliation(s)
| | | | | | | | | | - Peter Mohr
- Elbe Klinikum Buxtehude, Buxtehude, Germany
| | | | | | | | - Jacek Mackiewicz
- Institute of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Petr Arenberger
- Charles University Third Faculty of Medicine and University Hospital Královské Vinohrady, Prague, Czech Republic
| | | | - Tarek M Meniawy
- University of Western Australia and Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| |
Collapse
|
191
|
Bai X, Shaheen A, Grieco C, d’Arienzo PD, Mina F, Czapla JA, Lawless AR, Bongiovanni E, Santaniello U, Zappi H, Dulak D, Williamson A, Lee R, Gupta A, Li C, Si L, Ubaldi M, Yamazaki N, Ogata D, Johnson R, Park BC, Jung S, Madonna G, Hochherz J, Umeda Y, Nakamura Y, Gebhardt C, Festino L, Capone M, Ascierto PA, Johnson DB, Lo SN, Long GV, Menzies AM, Namikawa K, Mandala M, Guo J, Lorigan P, Najjar YG, Haydon A, Quaglino P, Boland GM, Sullivan RJ, Furness AJ, Plummer R, Flaherty KT. Dabrafenib plus trametinib versus anti-PD-1 monotherapy as adjuvant therapy in BRAF V600-mutant stage III melanoma after definitive surgery: a multicenter, retrospective cohort study. EClinicalMedicine 2023; 65:102290. [PMID: 37965433 PMCID: PMC10641479 DOI: 10.1016/j.eclinm.2023.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Both dabrafenib/trametinib (D/T) and anti-PD-1 monotherapy (PD-1) are approved adjuvant therapies for patients with stage III BRAF V600-mutant melanoma. However, there is still a lack of head-to-head comparative data. We aimed to describe efficacy and toxicity outcomes for these two standard therapies across melanoma centers. METHODS This multicenter, retrospective cohort study was conducted in 15 melanoma centers in Australia, China, Germany, Italy, Japan, UK, and US. We included adult patients with resected stage III BRAF V600-mutant melanoma who received either adjuvant D/T or PD-1 between Jul 2015 and Oct 2022. The primary endpoint was relapse-free survival (RFS). Secondary endpoints included overall survival (OS), recurrence pattern and toxicity. FINDINGS We included 598 patients with stage III BRAF V600-mutant melanoma who received either adjuvant D/T (n = 393 [66%]) or PD-1 (n = 205 [34%]) post definitive surgery between Jul 2015 and Oct 2022. At a median follow-up of 33 months (IQR 21-43), the median RFS was 51.0 months (95% CI 41.0-not reached [NR]) in the D/T group, significantly longer than PD-1 (44.8 months [95% CI 28.5-NR]) (univariate: HR 0.66, 95% CI 0.50-0.87, P = 0.003; multivariate: HR 0.58, 95% CI 0.39-0.86, P = 0.007), with comparable OS with PD-1 (multivariate, HR 0.90, 95% CI 0.48-1.70, P = 0.75). Similar findings were observed using a restricted-mean-survival-time model. Among those who experienced recurrence, the proportion of distant metastases was higher in the D/T cohort. D/T had a higher incidence of treatment modification due to adverse events (AEs) than PD-1, but fewer persistent AEs. INTERPRETATION In patients with stage III BRAF V600-mutant melanoma post definitive surgery, D/T yielded better RFS than PD-1, with higher transient but lower persistent toxicity, and comparable OS. D/T seems to provide a better outcome compared with PD-1, but a longer follow-up and ideally a large prospective trial are needed. FUNDING Dr. Xue Bai was supported by the Beijing Hospitals Authority Youth Programme (QMS20211101) for her efforts devoted to this study. Dr. Keith T. Flaherty was funded by Adelson Medical Research Foundation for the efforts devoted to this study.
Collapse
Affiliation(s)
- Xue Bai
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
- Massachusetts General Hospital, USA
| | | | | | | | - Florentia Mina
- Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | - Eleonora Bongiovanni
- Dermatologic Clinic, Department of Medical Sciences, University of Turin Medical School, Italy
| | - Umberto Santaniello
- Dermatologic Clinic, Department of Medical Sciences, University of Turin Medical School, Italy
| | | | - Dominika Dulak
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Rebecca Lee
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, UK
| | | | - Caili Li
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Si
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Rebecca Johnson
- Melanoma Institute Australia, The University of Sydney; Faculty of Medicine and Health, The University of Sydney; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Benjamin C. Park
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Seungyeon Jung
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabriele Madonna
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Juliane Hochherz
- Department of Dermatology, University Skin Cancer Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Yoshiyasu Umeda
- Department of Skin Oncology/Dermatology, Comprehensive Cancer Center, Saitama Medical University International Medical Center, Saitama, Japan
| | - Yasuhiro Nakamura
- Department of Skin Oncology/Dermatology, Comprehensive Cancer Center, Saitama Medical University International Medical Center, Saitama, Japan
| | - Christoffer Gebhardt
- Department of Dermatology, University Skin Cancer Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Lucia Festino
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Mariaelena Capone
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Paolo Antonio Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics - Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Serigne N. Lo
- Melanoma Institute Australia, The University of Sydney, North Sydney, NSW, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, The University of Sydney; Faculty of Medicine and Health, The University of Sydney; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Alexander M. Menzies
- Melanoma Institute Australia, The University of Sydney; Faculty of Medicine and Health, The University of Sydney; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Jun Guo
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, UK
| | | | | | - Pietro Quaglino
- Dermatologic Clinic, Department of Medical Sciences, University of Turin Medical School, Italy
| | | | | | | | | | | |
Collapse
|
192
|
Helvind NM, Brinch-Møller Weitemeyer M, Chakera AH, Hendel HW, Ellebæk E, Svane IM, Kjærskov MW, Bojesen S, Skyum H, Petersen SK, Bastholt L, Johansen C, Bidstrup PE, Hölmich LR. Stage-Specific Risk of Recurrence and Death From Melanoma in Denmark, 2008-2021: A National Observational Cohort Study of 25 720 Patients With Stage IA to IV Melanoma. JAMA Dermatol 2023; 159:1213-1222. [PMID: 37650576 PMCID: PMC10472263 DOI: 10.1001/jamadermatol.2023.3256] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023]
Abstract
Importance To ensure optimal treatment and surveillance of patients with melanoma, knowledge of the clinical stage-specific risk of recurrence, mortality, and recurrence patterns across the American Joint Committee on Cancer Eighth Edition (AJCC8) substages is needed. Objective To estimate stage-specific recurrence and melanoma-specific mortality rates, assess absolute stage-specific risks of recurrence and mortality, and describe stage-specific recurrence patterns, including conditional rates. Design Retrospective cohort study of prospectively collected nationwide population-based registry data. Setting Nationwide, population-based cohort study. Participants The 25 720 Danish patients, 18 years or older, diagnosed with first-time stage IA to IV cutaneous melanoma between January 1, 2008, and December 31, 2019, were included and followed up from time of primary treatment until December 31, 2021. Exposures First diagnosis of stage IA to IV cutaneous melanoma. Main Outcomes Stage-specific cumulative incidence of recurrence and melanoma-specific mortality, melanoma-specific recurrence-free survival, and assessed absolute stage-specific risks of recurrence and melanoma-specific mortality. Secondary outcomes were stage-specific recurrence patterns, including conditional rates, and melanoma-specific survival. Results We followed up 25 720 patients for a median of 5.9 years (95% CI, 58.9-59.3 years). Mean age was 59.1 years (95% CI, 58.9-59.3 years). Patients with stage IIB to IIC melanoma were older, had more comorbidities at diagnosis, and had the lowest rate of pathologic staging by sentinel node biopsy (81.6%-87.4%). A total of 10.6% of patients developed recurrence; first recurrence included distant recurrence, alone or with synchronous locoregional recurrence, in 56.6% of patients. We found a comparable risk of recurrence in stages IIIA and IIB (29.7% vs 33.2%) and in stages IIIB and IIC (35.9% vs 36.8%), respectively. Melanoma-specific mortality was comparable between stages IIIA and IIA (13.0% vs 13.6%) and between stages IIIB and IIB (18.4% vs 22.0%), respectively. These risk patterns persisted in cause-specific hazards models. Conclusions and Relevance This nationwide, population-based cohort study found that the increasing stages of the current AJCC8 staging system do not accurately reflect an increasing risk of recurrence and mortality in melanoma. The high proportion of distant recurrences suggests that hematogenous spread is a more common metastatic pathway than previously assumed, and surveillance with routine functional/cross-sectional imaging should be considered for stages IIB to IV. Future efforts should be put toward developing new tools for risk stratification and determining the survival effect of routine imaging in surveillance.
Collapse
Affiliation(s)
- Neel M. Helvind
- Department of Plastic Surgery, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| | | | - Annette H. Chakera
- Department of Plastic Surgery, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| | - Helle W. Hendel
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| | - Eva Ellebæk
- National Center for Cancer Immune Therapy, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
- Department of Oncology, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| | - Mette W. Kjærskov
- Department of General and Plastic Surgery, Vejle Hospital, Vejle, Denmark
| | - Sophie Bojesen
- Department of Plastic Surgery, Zealand University Hospital Roskilde, Roskilde, Denmark
| | - Helle Skyum
- Department of Plastic Surgery, Aalborg University Hospital, Aalborg, Denmark
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Christoffer Johansen
- Cancer Late Effect Research, Oncology Clinic, Center for Surgery and Cancer, Rigshospitalet, Copenhagen, Denmark
| | - Pernille E. Bidstrup
- Psychological Aspects of Cancer, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Lisbet R. Hölmich
- Department of Plastic Surgery, Copenhagen University Hospital: Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
193
|
Chen Y, Zhang J, Han G, Tang J, Guo F, Li W, Xie L, Xu H, Zhang X, Tian Y, Pan L, Shu Y, Ma L, Chen X. Efficacy and safety of XELOX combined with anlotinib and penpulimab vs XELOX as an adjuvant therapy for ctDNA-positive gastric and gastroesophageal junction adenocarcinoma: a protocol for a randomized, controlled, multicenter phase II clinical trial (EXPLORING study). Front Immunol 2023; 14:1232858. [PMID: 38022553 PMCID: PMC10644233 DOI: 10.3389/fimmu.2023.1232858] [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: 06/01/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Background The efficacy of current adjuvant chemotherapy for gastric adenocarcinoma/gastroesophageal junction adenocarcinoma (GA/GEJA) leaves much to be desired. ctDNA could serve as a potential marker to identify patients who are at higher risk of recurrence. Reinforcing standard adjuvant chemotherapy with immunotherapy has already been indicated to significantly improve clinical outcome, albeit such evidence is rare in GA/GEJA. Here, we intend to explore the clinical benefit of the reinforcement of adjuvant immunotherapy and antiangiogenics alongside with chemotherapy in patients who are deemed in high risk of recurrence by ctDNA analysis, which might shed light on further improvements in adjuvant therapy for GA/GEJA. Methods/Design This study is designed as a prospective, multicenter, randomized, controlled phase II study in patients histologically or cytologically diagnosed with GA/GEJA who underwent D2 gastrectomy and achieved R0 or R1 resection. From February 2022, a total of 300 stage III patients will be enrolled and subjected according to ctDNA sequencing results, and those with positive results will subsequently be randomized 1:1 to arm A or B. Patients in arm A will receive anlotinib, penpulimab and XELOX for 6-8 cycles, maintained with anlotinib and penpulimab for up to 1 year, while patients in arm B will receive XELOX alone for 6-8 cycles. ctDNA-negative patients will be assigned to arm C, and patients who are ctDNA positive but failed in randomization will be assigned to arm D. Patients in arms C and D will receive the investigator's choice of therapy. The primary endpoint is the median disease-free survival (DFS) of arm A versus arm B determined via CT/MRI imaging. Secondary endpoints include the DFS of ctDNA positive patients versus ctDNA negative patients, the 2- and 3-year DFS rates, overall survival (OS), the impact of hallmark molecules on the treatment response, adverse events (AEs), and the impact of nutrition status or exercise on recurrence. Discussion We expect that ctDNA would be a strong prognostic factor and ctDNA-positive patients are at higher risk of relapse than ctDNA-negative patients. The addition of anlotinib and penpulimab to XELOX, may contribute to delaying relapse in ctDNA-positive patients. Trial registration https://www.clinicaltrials.gov, identifier NCT05494060.
Collapse
Affiliation(s)
- Yizhang Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jiaguang Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gaohua Han
- Department of Oncology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Jie Tang
- Department of Oncology, Liyang People's Hospital, Changzhou, China
| | - Fen Guo
- Department of Oncology, Suzhou Municipal Hospital, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow, Suzhow, China
| | - Li Xie
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Xu
- Department of Gastric Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyi Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yitong Tian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lanlan Pan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Oncology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
194
|
Seth R, Agarwala SS, Messersmith H, Alluri KC, Ascierto PA, Atkins MB, Bollin K, Chacon M, Davis N, Faries MB, Funchain P, Gold JS, Guild S, Gyorki DE, Kaur V, Khushalani NI, Kirkwood JM, McQuade JL, Meyers MO, Provenzano A, Robert C, Santinami M, Sehdev A, Sondak VK, Spurrier G, Swami U, Truong TG, Tsai KK, van Akkooi A, Weber J. Systemic Therapy for Melanoma: ASCO Guideline Update. J Clin Oncol 2023; 41:4794-4820. [PMID: 37579248 DOI: 10.1200/jco.23.01136] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/09/2023] [Indexed: 08/16/2023] Open
Abstract
PURPOSE To provide guidance to clinicians regarding the use of systemic therapy for melanoma. METHODS American Society of Clinical Oncology convened an Expert Panel and conducted an updated systematic review of the literature. RESULTS The updated review identified 21 additional randomized trials. UPDATED RECOMMENDATIONS Neoadjuvant pembrolizumab was newly recommended for patients with resectable stage IIIB to IV cutaneous melanoma. For patients with resected cutaneous melanoma, adjuvant nivolumab or pembrolizumab was newly recommended for stage IIB-C disease and adjuvant nivolumab plus ipilimumab was added as a potential option for stage IV disease. For patients with unresectable or metastatic cutaneous melanoma, nivolumab plus relatlimab was added as a potential option regardless of BRAF mutation status and nivolumab plus ipilimumab followed by nivolumab was preferred over BRAF/MEK inhibitor therapy. Talimogene laherparepvec is no longer recommended as an option for patients with BRAF wild-type disease who have progressed on anti-PD-1 therapy. Ipilimumab- and ipilimumab-containing regimens are no longer recommended for patients with BRAF-mutated disease after progression on other therapies.This full update incorporates the new recommendations for uveal melanoma published in the 2022 Rapid Recommendation Update.Additional information is available at www.asco.org/melanoma-guidelines.
Collapse
Affiliation(s)
- Rahul Seth
- SUNY Upstate Medical University, Syracuse, NY
| | - Sanjiv S Agarwala
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | | | - Matias Chacon
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Nancy Davis
- Vanderbilt University Medical Center, Nashville, TN
| | - Mark B Faries
- The Angeles Clinic and Research Institute and Cedars Sinai Medical Center, Los Angeles, CA
| | | | | | | | | | | | | | - John M Kirkwood
- University of Pittsburgh School of Medicine and UPMC Hillman Cancer Institute, Pittsburgh, PA
| | | | - Michael O Meyers
- University of North Carolina School of Medicine, Chapel Hill, NC
| | | | - Caroline Robert
- Gustave Roussy Cancer Centre and Paris-Saclay University, Villejuif, France
| | - Mario Santinami
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Vernon K Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Katy K Tsai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Alexander van Akkooi
- Melanoma Institute Australia, University of Sydney and Royal Prince Alfred Hospital, Sydney, Australia
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY
| |
Collapse
|
195
|
Adeuyan O, Gordon ER, Kenchappa D, Bracero Y, Singh A, Espinoza G, Geskin LJ, Saenger YM. An update on methods for detection of prognostic and predictive biomarkers in melanoma. Front Cell Dev Biol 2023; 11:1290696. [PMID: 37900283 PMCID: PMC10611507 DOI: 10.3389/fcell.2023.1290696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/04/2023] [Indexed: 10/31/2023] Open
Abstract
The approval of immunotherapy for stage II-IV melanoma has underscored the need for improved immune-based predictive and prognostic biomarkers. For resectable stage II-III patients, adjuvant immunotherapy has proven clinical benefit, yet many patients experience significant adverse events and may not require therapy. In the metastatic setting, single agent immunotherapy cures many patients but, in some cases, more intensive combination therapies against specific molecular targets are required. Therefore, the establishment of additional biomarkers to determine a patient's disease outcome (i.e., prognostic) or response to treatment (i.e., predictive) is of utmost importance. Multiple methods ranging from gene expression profiling of bulk tissue, to spatial transcriptomics of single cells and artificial intelligence-based image analysis have been utilized to better characterize the immune microenvironment in melanoma to provide novel predictive and prognostic biomarkers. In this review, we will highlight the different techniques currently under investigation for the detection of prognostic and predictive immune biomarkers in melanoma.
Collapse
Affiliation(s)
- Oluwaseyi Adeuyan
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Emily R. Gordon
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Divya Kenchappa
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yadriel Bracero
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ajay Singh
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, United States
| | | |
Collapse
|
196
|
Huang J, Gong C, Zhou A. Modulation of gut microbiota: a novel approach to enhancing the effects of immune checkpoint inhibitors. Ther Adv Med Oncol 2023; 15:17588359231204854. [PMID: 37841750 PMCID: PMC10571694 DOI: 10.1177/17588359231204854] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have greatly improved the prognosis of some cancer patients, the majority still fail to respond adequately, and the available biomarkers cannot reliably predict drug efficacy. The gut microbiota has received widespread attention among the various intrinsic and extrinsic factors contributing to drug resistance. As an essential regulator of physiological function, the impact of gut microbiota on host immunity and response to cancer therapy is increasingly recognized. Several studies have demonstrated significant differences in gut microbiota between responders and nonresponders. The gut microbiota associated with better clinical outcomes is called 'favorable gut microbiota'. Significantly, interventions can alter the gut microbiota. By shifting the gut microbiota to the 'favorable' one through various modifications, preclinical and clinical studies have yielded more pronounced responses and better clinical outcomes when combined with ICIs treatment, providing novel approaches to improve the efficacy of cancer immunotherapy. These findings may be attributed to the effects of gut microbiota and its metabolites on the immune microenvironment and the systemic immune system, but the underlying mechanisms remain to be discovered. In this review, we summarize the clinical evidence that the gut microbiota is strongly associated with the outcomes of ICI treatment and describe the gut microbiota characteristics associated with better clinical outcomes. We then expand on the current prevalent modalities of gut microbiota regulation, provide a comprehensive overview of preclinical and clinical research advances in improving the therapeutic efficacy and prognosis of ICIs by modulating gut microbiota, and suggest fundamental questions we need to address and potential directions for future research expansion.
Collapse
Affiliation(s)
- Jinglong Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100020, China
| |
Collapse
|
197
|
Rhodin KE, Tyler DS, Zager JS, Beasley GM. Great Debate: Limb Infusion for Melanoma: A Thing of the Past? Ann Surg Oncol 2023; 30:6319-6324. [PMID: 37458946 DOI: 10.1245/s10434-023-13765-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/04/2023] [Indexed: 09/20/2023]
Affiliation(s)
- Kristen E Rhodin
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Douglas S Tyler
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
198
|
Jasper S, Keim U, Leiter U, Amaral T, Flatz L, Forschner A. Die Prognose des Melanoms im Kopf-Hals-Bereich im Stadium II hängt vom histologischen Subtyp ab. J Dtsch Dermatol Ges 2023; 21:1137-1147. [PMID: 37845056 DOI: 10.1111/ddg.15164_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 05/28/2023] [Indexed: 10/18/2023]
Abstract
ZusammenfassungHintergrund und ZieleDie Melanom‐Leitlinie basiert hauptsächlich auf dem AJCC‐Stadium. Hierbei wird nicht zwischen den histologischen Subtypen wie dem superfiziell spreitenden Melanom (SSM), dem Lentigo‐maligna‐Melanom (LMM) oder dem nodulären malignen Melanom (NM) unterschieden. Ziel der Studie war es zu untersuchen, ob sich Patienten mit LMM im klinischen Verlauf von Patienten mit SSM/NM unterscheiden. Dies ist aktuell besonders wichtig, da die adjuvante Anti‐PD‐1‐Therapie für Melanome im Stadium IIB und IIC zugelassen wurde.Patienten und MethodikDie Daten wurden aus dem Zentralregister „malignes Melanom“ entnommen. Es wurden nur Patienten mit LMM, SSM oder NM des Kopf‐Hals‐Bereichs und Primärdiagnose zwischen dem 01.01.2000 und dem 31.12.2019 eingeschlossen. Das progressionsfreie Überleben (PFÜ), das melanomspezifische Überleben (MSÜ) und das Metastasierungsmuster wurden für die Gruppe der LMM im Vergleich zur Gruppe der SSM/NM analysiert.ErgebnisseDie LMM‐Kohorte (n = 902) hatte ein signifikant besseres MSÜ als die SSM/NM‐Kohorte (n = 604). Beim PFÜ gab es keinen Unterschied. Das 5‐Jahres‐MSÜ der LMM‐Kohorte im Stadium II betrug 88,5 % (95 % KI 81,4–95,6), im Vergleich dazu das der SSM/NM‐Kohorte im Stadium II 79,7 % (95 % KI 72,8–86,6).SchlussfolgerungEs scheint nicht angebracht zu sein, eine adjuvante Therapie bei LMM‐Patienten im Stadium II im gleichen Umfang durchzuführen, wie bei Patienten mit SSM/NM.
Collapse
Affiliation(s)
- Sophie Jasper
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| | - Ulrike Keim
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| | - Ulrike Leiter
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| | - Teresa Amaral
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| | - Lukas Flatz
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| | - Andrea Forschner
- Abteilung für Dermatologie, Zentrum für Dermatoonkologie, Universitätsklinikum Tübingen
| |
Collapse
|
199
|
Rhodin KE, Jung SH, Elleson K, DePalo D, Straker R, McKinley S, Beekman K, Parker L, Chen S, Iyer MK, Salama AKS, Bartlett E, Karakousis G, Zager JS, Tyler DS, Beasley GM. Timing of Adjuvant Immunotherapy in Stage III Melanoma, Does it Matter? Ann Surg Oncol 2023; 30:6340-6352. [PMID: 37481487 PMCID: PMC10530114 DOI: 10.1245/s10434-023-13935-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND The optimal time to initiate adjuvant immune checkpoint inhibitors (ICI) following resection remains undefined. Herein, we investigated the impact of time to adjuvant ICI on survival in patients with stage III melanoma. METHODS Patients with resected stage III melanoma receiving adjuvant immune therapy were identified within a multi-institutional retrospective cohort. Patients were stratified by time to adjuvant ICI: within 6 weeks, 6-12 weeks, and greater than 12 weeks from surgery. Recurrence-free survival (RFS) was compared among time strata with Kaplan-Meier and Cox proportional hazards methods in the multi-institutional cohort. RESULTS Altogether, 626 patients were identified within the multi-institutional cohort: 39% of patients initiated adjuvant ICI within 6 weeks, 42.2% within 6-12 weeks, and 18.8% greater than 12 weeks from surgery. In a multivariate Cox model, adjusting for histology, nodal tumor burden, and pathologic stage, we found that increased time to adjuvant ICI was associated with improved RFS. Patients who initiated adjuvant ICI within 6 weeks of surgery had worse RFS. These findings were preserved in a conditional landmark analysis and separate subgroups of patients with (1) new melanoma diagnoses, (2) occult stage III disease, and (3) those receiving anti-PD-1 monotherapy. CONCLUSIONS Outcomes for patients with stage III melanoma are not compromised when adjuvant ICI is initiated beyond 6 weeks from resection. Additional work is needed to better understand the underlying mechanisms and implications of timing of adjuvant ICI on long-term outcomes.
Collapse
Affiliation(s)
| | - Sin-Ho Jung
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Kelly Elleson
- Department of Surgery, Moffitt Cancer Center, Tampa, FL, USA
| | - Danielle DePalo
- Department of Surgery, Moffitt Cancer Center, Tampa, FL, USA
| | - Richard Straker
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia McKinley
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kate Beekman
- Department of Surgery, Moffitt Cancer Center, Tampa, FL, USA
| | - Lily Parker
- Department of Surgery, Moffitt Cancer Center, Tampa, FL, USA
| | - Suephy Chen
- Department of Dermatology, Duke University, Durham, NC, USA
| | | | | | - Edmund Bartlett
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giorgos Karakousis
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Douglas S Tyler
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
200
|
Rhodin KE, Beasley GM. ASO Author Reflections: It's About Time? Ann Surg Oncol 2023; 30:6353-6354. [PMID: 37481491 DOI: 10.1245/s10434-023-13960-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/24/2023]
Affiliation(s)
- Kristen E Rhodin
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Georgia M Beasley
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|