201
|
Larkin J, Del Vecchio M, Mandalá M, Gogas H, Arance Fernandez AM, Dalle S, Cowey CL, Schenker M, Grob JJ, Chiarion-Sileni V, Marquez-Rodas I, Butler MO, Di Giacomo AM, Middleton MR, Lutzky J, de la Cruz-Merino L, Arenberger P, Atkinson V, Hill AG, Fecher LA, Millward M, Nathan PD, Khushalani NI, Queirolo P, Ritchings C, Lobo M, Askelson M, Tang H, Dolfi S, Ascierto PA, Weber J. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III/IV Melanoma: 5-Year Efficacy and Biomarker Results from CheckMate 238. Clin Cancer Res 2023; 29:3352-3361. [PMID: 37058595 PMCID: PMC10472092 DOI: 10.1158/1078-0432.ccr-22-3145] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 04/16/2023]
Abstract
PURPOSE In the phase III CheckMate 238 study, adjuvant nivolumab significantly improved recurrence-free survival (RFS) and distant metastasis-free survival versus ipilimumab in patients with resected stage IIIB-C or stage IV melanoma, with benefit sustained at 4 years. We report updated 5-year efficacy and biomarker findings. PATIENTS AND METHODS Patients with resected stage IIIB-C/IV melanoma were stratified by stage and baseline programmed death cell ligand 1 (PD-L1) expression and received nivolumab 3 mg/kg every 2 weeks or ipilimumab 10 mg/kg every 3 weeks for four doses and then every 12 weeks, both intravenously for 1 year until disease recurrence, unacceptable toxicity, or withdrawal of consent. The primary endpoint was RFS. RESULTS At a minimum follow-up of 62 months, RFS with nivolumab remained superior to ipilimumab (HR = 0.72; 95% confidence interval, 0.60-0.86; 5-year rates of 50% vs. 39%). Five-year distant metastasis-free survival (DMFS) rates were 58% with nivolumab versus 51% with ipilimumab. Five-year overall survival (OS) rates were 76% with nivolumab and 72% with ipilimumab (75% data maturity: 228 of 302 planned events). Higher levels of tumor mutational burden (TMB), tumor PD-L1, intratumoral CD8+ T cells and IFNγ-associated gene expression signature, and lower levels of peripheral serum C-reactive protein were associated with improved RFS and OS with both nivolumab and ipilimumab, albeit with limited clinically meaningful predictive value. CONCLUSIONS Nivolumab is a proven adjuvant treatment for resected melanoma at high risk of recurrence, with sustained, long-term improvement in RFS and DMFS compared with ipilimumab and high OS rates. Identification of additional biomarkers is needed to better predict treatment outcome. See related commentary by Augustin and Luke, p. 3253.
Collapse
Affiliation(s)
- James Larkin
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | | | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | - Ivan Marquez-Rodas
- General University Hospital Gregorio Marañón and CIBERONC, Madrid, Spain
| | | | | | | | - Jose Lutzky
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Luis de la Cruz-Merino
- Hospital Universitario Virgen Macarena, Clinical Oncology Department, University of Seville, Seville, Spain
| | - Petr Arenberger
- Charles University Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Victoria Atkinson
- Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
| | | | | | - Michael Millward
- University of Western Australia and Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | | | | | | | | | | | | | - Hao Tang
- Bristol Myers Squibb, Princeton, New Jersey
| | | | - Paolo A. Ascierto
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York
| |
Collapse
|
202
|
Vuoristo M, Juteau S, Koljonen V, Hernberg M, Mätzke S, Ilmonen S, Jahkola T. Hot dots - which nodes should be removed in sentinel lymph node biopsy for melanoma? Acta Oncol 2023; 62:1021-1027. [PMID: 37493624 DOI: 10.1080/0284186x.2023.2238558] [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: 05/07/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND Sentinel lymph node biopsy (SLNB) is a critical staging tool for melanoma patients. The optimal number of lymph nodes removed in SLNB remains unclear. In this study, we retrospectively analysed and tested different criteria for selecting sentinel lymph nodes (SLNs) by radiotracer uptake and blue dye, and their impact on nodal staging. We also evaluated the association between SLN tumour burden and radiotracer uptake. METHODS The study population consisted of melanoma patients undergoing SLNB. During the operation all radioactive and blue nodes were removed and sent for histopathological analysis. The ex vivo radioactive count and presence of blue dye of each node were recorded, and these were correlated with presence and size of metastasis in each SLN. RESULTS Altogether 175 patients with clinically occult metastasis presented with one or more positive, i.e. metastatic, SLNs. The mean number of lymph nodes removed was 4.5, and the mean number of positive lymph nodes was 1.5 per patient. The most radioactive or hottest node was negative in 38 patients (22%). By removing the hottest node and all nodes with radioactivity >10% of the hottest node, 97% of patients would have been staged correctly. In five patients, metastasis was found solely in a SLN with radioactivity <10% of the hottest node. Of all 267 positive nodes removed, 125 (47%) contained blue dye. Patients with a negative hottest node were associated with lower SLN tumour burden. CONCLUSIONS By removing the hottest node and all nodes with radioactivity >10% of the hottest node, 97% of patients with SLN metastases are correctly staged with or without using blue dye.
Collapse
Affiliation(s)
- Mikko Vuoristo
- Department of Plastic Surgery, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Susanna Juteau
- Department of Pathology, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Micaela Hernberg
- Department of Oncology, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Sorjo Mätzke
- Department of Clinical Physiology and Nuclear Medicine, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Suvi Ilmonen
- Department of Plastic Surgery, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Tiina Jahkola
- Department of Plastic Surgery, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
203
|
Dar N, Gradecki SE, Gaughan EM. Case Report: Dynamic overlap of melanoma, sarcoidosis, and targeted therapy for BRAF-mutant melanoma. Front Oncol 2023; 13:1217179. [PMID: 37706179 PMCID: PMC10495986 DOI: 10.3389/fonc.2023.1217179] [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: 05/04/2023] [Accepted: 07/31/2023] [Indexed: 09/15/2023] Open
Abstract
Targeted therapies, including BRAF and MEK inhibitors, are valuable treatment options for patients with unresectable or metastatic BRAF V600-mutant melanoma. With the improvement in survival seen with modern melanoma therapeutics, clinicians are learning the variable patterns associated with extended clinical courses. Sarcoidosis is characterized by non-caseating granulomatous inflammation of unknown etiology, often presenting with cutaneous, lung, or lymph node involvement. There is a known association between sarcoidosis and melanoma, and sarcoidosis is increasingly seen and described in the setting of anti-melanoma therapy. The challenge for clinicians is to differentiate between sarcoid-related and malignancy-related findings, which may follow a variable course over years. We present two cases of BRAF and MEK inhibitor-related sarcoidosis in patients with melanoma and review the literature. The dynamic nature of the clinical and radiographic findings impacted patient management and clinical decisions for years of their treatment course.
Collapse
Affiliation(s)
- Nakul Dar
- University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sarah E. Gradecki
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Elizabeth M. Gaughan
- Division of Hematology and Medical Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
204
|
Bye BA, Jack J, Pierce A, Walsh RM, Eades A, Chalise P, Olou A, VanSaun MN. Combined PI3K and MAPK inhibition synergizes to suppress PDAC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553438. [PMID: 37645960 PMCID: PMC10462031 DOI: 10.1101/2023.08.15.553438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Oncogenic KRAS mutations are nearly ubiquitous in pancreatic ductal adenocarcinoma (PDAC), yet therapeutic attempts to target KRAS as well as its target MAPK pathway effectors have shown limited success due to the difficulty to pharmacologically target KRAS, inherent drug resistance in PDAC cells, and acquired resistance through activation of alternative mitogenic pathways such JAK-STAT and PI3K-AKT. While KRAS canonically drives the MAPK signaling pathway via RAF-MEK-ERK, it is also known to play a role in PI3K-AKT signaling. Our therapeutic study targeted the PI3K-AKT pathway with the drug Omipalisib (p110α/β/δ/γ and mTORC1/2 inhibitor) in combination with MAPK pathway targeting drug Trametinib (MEK1/2 inhibitor) or SHP099-HCL (SHP099), which is an inhibitor of the KRAS effector SHP2. Western blot analysis demonstrated that application of Trametinib or SHP099 alone selectively blocked ERK phosphorylation (pERK) but failed to suppress phosphorylated AKT (pAKT) and in some instances increased pAKT levels. Conversely, Omipalisib alone successfully inhibited pAKT but failed to suppress pERK. Therefore, we hypothesized that a combination therapeutic comprised of Omipalisib with either Trametinib or SHP099 would inhibit two prominent mitogenic pathways, MEK and PI3K-AKT, to more effectively suppress pancreatic cancer. In vitro studies demonstrated that both Omipalisib/Trametinib and Omipalisib/SHP099 combination therapeutic strategies were generally more effective than treatment with each drug individually at reducing proliferation, colony formation, and cell migration compared to vehicle controls. Additionally, we found that while combination Omipalisib/SHP099 treatment reduced implanted tumor growth in vivo , the Omipalisib/Trametinib treatment was significantly more effective. Therefore, we additionally tested the Omipalisib/Trametinib combination therapeutic in the highly aggressive PKT (Ptf1a cre , LSL-Kras G12D , TGFbR2 fl/fl ) spontaneous mouse model of PDAC. We subsequently found that PKT mice treated with the Omipalisib/Trametinib combination therapeutic survived significantly longer than mice treated with either drug alone, and more than doubled the mean survival time of vehicle control mice. Altogether, our data support the importance of a dual treatment strategy targeting both MAPK and PI3K-AKT pathways.
Collapse
|
205
|
Eljilany I, Castellano E, Tarhini AA. Adjuvant Therapy for High-Risk Melanoma: An In-Depth Examination of the State of the Field. Cancers (Basel) 2023; 15:4125. [PMID: 37627153 PMCID: PMC10453009 DOI: 10.3390/cancers15164125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The consideration of systemic adjuvant therapy is recommended for patients with stage IIB-IV melanoma who have undergone surgical resection due to a heightened risk of experiencing melanoma relapse and mortality from melanoma. Adjuvant therapy options tested over the past three decades include high-dose interferon-α, immune checkpoint inhibitors (pembrolizumab, nivolumab), targeted therapy (dabrafenib-trametinib for BRAF mutant melanoma), radiotherapy and chemotherapy. Most of these therapies have been demonstrated to enhance relapse-free survival (RFS) but with limited to no impact on overall survival (OS), as reported in randomized trials. In contemporary clinical practice, the adjuvant treatment approach for surgically resected stage III-IV melanoma has undergone a notable shift towards the utilization of nivolumab, pembrolizumab, and BRAF-MEK inhibitors, such as dabrafenib plus trametinib (specifically for BRAF mutant melanoma) due to the significant enhancements in RFS observed with these treatments. Pembrolizumab has obtained regulatory approval in the United States to treat resected stage IIB-IIC melanoma, while nivolumab is currently under review for the same indication. This review comprehensively analyzes completed phase III adjuvant therapy trials in adjuvant therapy. Additionally, it provides a summary of ongoing trials and an overview of the main challenges and future directions with adjuvant therapy.
Collapse
Affiliation(s)
- Islam Eljilany
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ella Castellano
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Ahmad A. Tarhini
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
206
|
Mazzera L, Abeltino M, Lombardi G, Cantoni AM, Jottini S, Corradi A, Ricca M, Rossetti E, Armando F, Peli A, Ferrari A, Martinelli G, Scupoli MT, Visco C, Bonifacio M, Ripamonti A, Gambacorti-Passerini C, Bonati A, Perris R, Lunghi P. MEK1/2 regulate normal BCR and ABL1 tumor-suppressor functions to dictate ATO response in TKI-resistant Ph+ leukemia. Leukemia 2023; 37:1671-1685. [PMID: 37386079 PMCID: PMC10400427 DOI: 10.1038/s41375-023-01940-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 07/01/2023]
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) remains a clinical challenge in Ph-positive variants of chronic myeloid leukemia. We provide mechanistic insights into a previously undisclosed MEK1/2/BCR::ABL1/BCR/ABL1-driven signaling loop that may determine the efficacy of arsenic trioxide (ATO) in TKI-resistant leukemic patients. We find that activated MEK1/2 assemble into a pentameric complex with BCR::ABL1, BCR and ABL1 to induce phosphorylation of BCR and BCR::ABL1 at Tyr360 and Tyr177, and ABL1, at Thr735 and Tyr412 residues thus provoking loss of BCR's tumor-suppression functions, enhanced oncogenic activity of BCR::ABL1, cytoplasmic retention of ABL1 and consequently drug resistance. Coherently, pharmacological blockade of MEK1/2 induces dissociation of the pentameric MEK1/2/BCR::ABL1/BCR/ABL1 complex and causes a concurrent BCRY360/Y177, BCR::ABL1Y360/Y177 and cytoplasmic ABL1Y412/T735 dephosphorylation thereby provoking the rescue of the BCR's anti-oncogenic activities, nuclear accumulation of ABL1 with tumor-suppressive functions and consequently, growth inhibition of the leukemic cells and an ATO sensitization via BCR-MYC and ABL1-p73 signaling axes activation. Additionally, the allosteric activation of nuclear ABL1 was consistently found to enhance the anti-leukemic effects of the MEK1/2 inhibitor Mirdametinib, which when combined with ATO, significantly prolonged the survival of mice bearing BCR::ABL1-T315I-induced leukemia. These findings highlight the therapeutic potential of MEK1/2-inhibitors/ATO combination for the treatment of TKI-resistant leukemia.
Collapse
Affiliation(s)
- Laura Mazzera
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuela Abeltino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | | | - Stefano Jottini
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Attilio Corradi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Micaela Ricca
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | - Elena Rossetti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- National Healthcare Service (SSN-Servizio Sanitario Nazionale) ASL Piacenza, Piacenza, Italy
| | - Federico Armando
- Department of Veterinary Science, University of Parma, Parma, Italy
- University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Angelo Peli
- Department for Life Quality Studies Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Ferrari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
- Institute of Hematology "L. e A. Seragnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlo Visco
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Massimiliano Bonifacio
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Alessia Ripamonti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Antonio Bonati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberto Perris
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy
| | - Paolo Lunghi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy.
| |
Collapse
|
207
|
Li AT, Law J, Ch'ng S, Scolyer RA, Thompson JF, Lo SN, Varey AHR. External validation in an Australian population of the EORTC-DeCOG nomogram predicting recurrence, distant metastasis and overall mortality in melanoma patients with positive sentinel lymph nodes. Eur J Cancer 2023; 189:112901. [PMID: 37263897 DOI: 10.1016/j.ejca.2023.04.017] [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: 12/15/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Calculating an accurate prognosis for melanoma patients who have a positive sentinel node (SN) biopsy is important both for them and for their treating doctors to guide decision-making, particularly when adjuvant systemic therapy is being considered. The recently published EORTC-DeCOG nomograms aim to provide this via an online portal that predicts 5-year rates for recurrence, distant metastasis and overall mortality. The present study provides external validation of these nomograms. METHODS/MATERIALS De-identified data from patients with a positive SN biopsy between 2003 and 2015 were extracted from the prospectively maintained Melanoma Institute Australia (MIA) research database. ROC-curves with C-statistics, regression co-efficients and Decision Curve Net Benefit analyses were performed using the integrated private validation portal on the nomograms' hosting platform (Evidencio). RESULTS Complete data were available for 352 patients. The respective C-statistics for recurrence, distant metastasis and overall mortality nomogram validations were 0.68, 0.69 and 0.66. CONCLUSION The performance of the nomograms in predicting recurrence and distant metastasis was similar in the MIA and the development populations, suggesting that they are robust. However, the overall mortality nomogram performance was significantly poorer in the MIA population (C-statistic 0.66) than in the original EORTC-DeCOG derivation cohort (C-statistic 0.70) and may therefore be less reliable for clinical use.
Collapse
Affiliation(s)
- Andrew T Li
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
| | - Jenaleen Law
- Department of Plastic & Reconstructive Surgery, Westmead Hospital, Sydney, NSW, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia; Department of Plastic Surgery, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexander H R Varey
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine & Health, The University of Sydney, Sydney, NSW, Australia; Department of Plastic & Reconstructive Surgery, Westmead Hospital, Sydney, NSW, Australia.
| |
Collapse
|
208
|
Lee R, Mandala M, Long GV, Eggermont AMM, van Akkooi ACJ, Sandhu S, Garbe C, Lorigan P. Adjuvant therapy for stage II melanoma: the need for further studies. Eur J Cancer 2023; 189:112914. [PMID: 37301717 DOI: 10.1016/j.ejca.2023.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 06/12/2023]
Abstract
Immunotherapy with checkpoint inhibitors has revolutionised the outcomes for melanoma patients. In the metastatic setting, patients treated with nivolumab and ipilimumab have an expected 5-year survival of> 50%. For patients with resected high-risk stage III disease, adjuvant pembrolizumab, nivolumab or dabrafenib and trametinib are associated with a significant improvement in both relapse-free survival (RFS) and distant metastasis-free survival (DMFS). More recently neoadjuvant immunotherapy has shown very promising outcomes in patients with clinically detectable nodal disease and is likely to become a new standard of care. For stage IIB/C disease, two pivotal adjuvant trials of pembrolizumab and nivolumab have also reported a significant improvement in both RFS and DMFS. However, the absolute benefit is low and there are concerns about the risk of severe toxicities as well as long-term morbidity from endocrine toxicity. Ongoing registration phase III trials are currently evaluating newer immunotherapy combinations and the role of BRAF/MEK-directed targeted therapy for stage II melanoma. However, our ability to personalise therapy based on molecular risk stratification has lagged behind the development of novel immune therapies. There is a critical need to evaluate the use of tissue and blood-based biomarkers, to better select patients that will recur and avoid unnecessary treatment for the majority of patients cured by surgery alone.
Collapse
Affiliation(s)
- Rebecca Lee
- The Christie NHS Foundation Trust, Department of Medical Oncology, Manchester, UK; The University of Manchester, Division of Cancer Sciences, Manchester, UK
| | - Mario Mandala
- University of Perugia, Perugia, Italy; Ospedale Papa Givoanni XXIII, Bergamo, Italy
| | - Georgina V Long
- Melanoma Institute Australia, Faculty of Medicine and Health, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Alexander M M Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, the Netherlands; Comprehensive Cancer Center München, Technical University München & Ludwig Maximiliaan University, München, Germany
| | - Alexander C J van Akkooi
- Comprehensive Cancer Center München, Technical University München & Ludwig Maximiliaan University, München, Germany; Melanoma Institute Australia, Faculty of Medicine and Health, The University of Sydney, Royal Prince Alfred Hospital, Sydney, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
| | - Claus Garbe
- Centre for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Department of Medical Oncology, Manchester, UK; The University of Manchester, Division of Cancer Sciences, Manchester, UK.
| |
Collapse
|
209
|
Long GV, Swetter SM, Menzies AM, Gershenwald JE, Scolyer RA. Cutaneous melanoma. Lancet 2023:S0140-6736(23)00821-8. [PMID: 37499671 DOI: 10.1016/s0140-6736(23)00821-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/17/2023] [Accepted: 04/19/2023] [Indexed: 07/29/2023]
Abstract
Cutaneous melanoma is a malignancy arising from melanocytes of the skin. Incidence rates are rising, particularly in White populations. Cutaneous melanoma is typically driven by exposure to ultraviolet radiation from natural sunlight and indoor tanning, although there are several subtypes that are not related to ultraviolet radiation exposure. Primary melanomas are often darkly pigmented, but can be amelanotic, with diagnosis based on a combination of clinical and histopathological findings. Primary melanoma is treated with wide excision, with margins determined by tumour thickness. Further treatment depends on the disease stage (following histopathological examination and, where appropriate, sentinel lymph node biopsy) and can include surgery, checkpoint immunotherapy, targeted therapy, or radiotherapy. Systemic drug therapies are recommended as an adjunct to surgery in patients with resectable locoregional metastases and are the mainstay of treatment in advanced melanoma. Management of advanced melanoma is complex, particularly in those with cerebral metastasis. Multidisciplinary care is essential. Systemic drug therapies, particularly immune checkpoint inhibitors, have substantially increased melanoma survival following a series of landmark approvals from 2011 onward.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia.
| | - Susan M Swetter
- Department of Dermatology and Pigmented Lesion and Melanoma Program, Stanford University Medical Center and Cancer Institute, Stanford, CA, USA; Department of Dermatology, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Mater Hospital, Sydney, NSW, Australia
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology and Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; NSW Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
210
|
Lucci A, Addanki S, Chiang YJ, Meas S, Sarli VN, Upshaw JR, Manchem M, Patel SP, Wargo JA, Gershenwald JE, Ross MI. Presence of Circulating Tumor Cells Predates Imaging Detection of Relapse in Patients with Stage III Melanoma. Cancers (Basel) 2023; 15:3630. [PMID: 37509290 PMCID: PMC10377914 DOI: 10.3390/cancers15143630] [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] [Received: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Stage III melanoma includes nodal metastasis or in-transit disease. Five-year survival rates vary between 32% and 93%. The identification of high-risk patients is important for clinical decision making. We demonstrated previously that ≥1 circulating tumor cells (CTCs) at baseline was associated with recurrence. In this study, we investigated how frequently CTCs were identified prior to radiologically detected recurrence. Stage III patients (n = 325) had imaging at baseline and q 3 months. Baseline and q 6-12 months blood draws (7.5 mL) were performed to identify CTCs up to 3.5 years from diagnosis. CTC assessment was performed using the immunomagnetic capture of CD146-positive cells and anti-MEL-PE. The presence of one or more CTCs was considered positive. We analyzed the cohort of patients with relapse confirmed by radiologic imaging. CTC collection dates were assessed to determine the lead time for CTC detection. CTC-negative patients were significantly less likely to relapse compared to patients positive for CTCs (p-value < 0.001). Within the 325-patient cohort, 143 patients (44%) had recurrence, with a median follow-up of 52 months from diagnosis. The cohort (n = 143) with positive imaging and CTC results revealed 76% of patients (108/143) had CTC+ results before the radiological identification of relapse. The median time between positive CTC and positive imaging was 9 months. CTCs were positive in >75% of patients prior to relapse at a median of 9 months before radiologic detection.
Collapse
Affiliation(s)
- Anthony Lucci
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sridevi Addanki
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi-Ju Chiang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Salyna Meas
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanessa N Sarli
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joshua R Upshaw
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mayank Manchem
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
211
|
Mooradian MJ, Sullivan RJ. The case for adjuvant BRAF-targeted therapy versus adjuvant anti-PD-1 therapy for patients with resected, high-risk melanoma. Cancer 2023; 129:2117-2121. [PMID: 37345669 DOI: 10.1002/cncr.34806] [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: 01/17/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 06/23/2023]
Abstract
The development of highly effective BRAF-targeted therapy and immune checkpoint inhibition for patients with advanced metastatic melanoma has transformed the treatment of this disease. More recently, these advances have moved into the resected, high-risk stage II and III settings. For patients with resected, BRAF-mutant stage III melanoma, there are no head-to-head data to support the use of BRAF-targeted therapy (specifically the combination of dabrafenib and trametinib) with either single-agent nivolumab or pembrolizumab. Because the relapse-free and distant metastasis-free survivals are similar in a cross-trial comparison, it is not clear what the best option for these patients is. In this piece, the authors argue on behalf of and against both approaches. PLAIN LANGUAGE SUMMARY: Two types of therapy exist for patients diagnosed with melanoma who have completed surgery and remain at high risk for tumor recurrence: (1) drugs that target the BRAF mutation (found in ∼50% of patients) and (2) immunotherapy. There are no data showing that either approach is better than the other, so the choice of which therapy is best for an individual patient can be challenging. In this article, we make arguments for and against each option.
Collapse
Affiliation(s)
- Meghan J Mooradian
- Division of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
212
|
Rutkowski P, Czarnecka AM. Pembrolizumab for the adjuvant treatment of IIB or IIC melanoma. Expert Rev Anticancer Ther 2023; 23:897-902. [PMID: 37573515 DOI: 10.1080/14737140.2023.2247565] [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: 12/04/2022] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Up to 30% of patients with stage IIB and 50% of stage IIC melanoma experience recurrence within 5 years after radical surgery. Adjuvant treatment is expected to improve this prognosis. AREAS COVERED Pembrolizumab (MK-3475) is a humanized monoclonal antibody that acts against the programmed cell death 1 (PD-1) receptor. Pembrolizumab was first approved in monotherapy for the treatment of unresectable/metastatic melanoma based on the results of the prospective KEYNOTE-001, KEYNOTE-002, and KEYNOTE-006 trials. KEYNOTE-716 is the randomized phase III trial of pembrolizumab treatment in resected stage II melanoma. Treatment with pembrolizumab is statistically significant, reducing the risk of recurrence as well as distant metastases risk after primary tumor resection. Pembrolizumab treatment has a 24-month RFS rate of 81.2% (HR 0.64 vs placebo) and a DMFS rate of 88.1%. EXPERT OPINION 1-year adjuvant pembrolizumab treatment of stage IIB/C melanoma patients significantly reduces recurrence or death risk. The safety profile of adjuvant treatment is not different from previously reported and is manageable. Longer follow-up is required to fully understand the efficacy and safety of adjuvant therapy for stage II melanoma, as the number of patients needed to treat is twice as high as for stage III patients.
Collapse
Affiliation(s)
- Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
213
|
Jansen P, Baguer DO, Duschner N, Arrastia JL, Schmidt M, Landsberg J, Wenzel J, Schadendorf D, Hadaschik E, Maass P, Schaller J, Griewank KG. Deep learning detection of melanoma metastases in lymph nodes. Eur J Cancer 2023; 188:161-170. [PMID: 37257277 DOI: 10.1016/j.ejca.2023.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND In melanoma patients, surgical excision of the first draining lymph node, the sentinel lymph node (SLN), is a routine procedure to evaluate lymphogenic metastases. Metastasis detection by histopathological analysis assesses multiple tissue levels with hematoxylin and eosin and immunohistochemically stained glass slides. Considering the amount of tissue to analyze, the detection of metastasis can be highly time-consuming for pathologists. The application of artificial intelligence in the clinical routine has constantly increased over the past few years. METHODS In this multi-center study, a deep learning method was established on histological tissue sections of sentinel lymph nodes collected from the clinical routine. The algorithm was trained to highlight potential melanoma metastases for further review by pathologists, without relying on supplementary immunohistochemical stainings (e.g. anti-S100, anti-MelanA). RESULTS The established method was able to detect the existence of metastasis on individual tissue cuts with an area under the curve of 0.9630 and 0.9856 respectively on two test cohorts from different laboratories. The method was able to accurately identify tumour deposits>0.1 mm and, by automatic tumour diameter measurement, classify these into 0.1 mm to -1.0 mm and>1.0 mm groups, thus identifying and classifying metastasis currently relevant for assessing prognosis and stratifying treatment. CONCLUSIONS Our results demonstrate that AI-based SLN melanoma metastasis detection has great potential and could become a routinely applied aid for pathologists. Our current study focused on assessing established parameters; however, larger future AI-based studies could identify novel biomarkers potentially further improving SLN-based prognostic and therapeutic predictions for affected patients.
Collapse
Affiliation(s)
- Philipp Jansen
- Department of Dermatology, University Hospital Bonn, Bonn 53127, Germany
| | | | | | | | | | - Jennifer Landsberg
- Department of Dermatology, University Hospital Bonn, Bonn 53127, Germany
| | - Jörg Wenzel
- Department of Dermatology, University Hospital Bonn, Bonn 53127, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen 45147, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, Essen 45147, Germany
| | | | - Jörg Schaller
- Dermatopathologie Duisburg Essen GmbH, Essen 45329, Germany
| | - Klaus Georg Griewank
- Department of Dermatology, University Hospital Essen, Essen 45147, Germany; Dermatopathologie bei Mainz, Nieder-Olm 55268, Germany.
| |
Collapse
|
214
|
Dasyam N, Sharples KJ, Barrow C, Huang Y, Bauer E, Mester B, Wood CE, Authier-Hall A, Dzhelali M, Ostapowicz T, Kumar R, Lowe J, Maxwell A, Burn OK, Williams GM, Carley SE, Caygill G, Jones J, Chan STS, Hinder VA, Macapagal J, McCusker M, Weinkove R, Brimble MA, Painter GF, Findlay MP, Dunbar PR, Gasser O, Hermans IF. A randomised controlled trial of long NY-ESO-1 peptide-pulsed autologous dendritic cells with or without alpha-galactosylceramide in high-risk melanoma. Cancer Immunol Immunother 2023; 72:2267-2282. [PMID: 36881133 PMCID: PMC10264280 DOI: 10.1007/s00262-023-03400-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023]
Abstract
AIM We have previously reported that polyfunctional T cell responses can be induced to the cancer testis antigen NY-ESO-1 in melanoma patients injected with mature autologous monocyte-derived dendritic cells (DCs) loaded with long NY-ESO-1-derived peptides together with α-galactosylceramide (α-GalCer), an agonist for type 1 Natural Killer T (NKT) cells. OBJECTIVE To assess whether inclusion of α-GalCer in autologous NY-ESO-1 long peptide-pulsed DC vaccines (DCV + α-GalCer) improves T cell responses when compared to peptide-pulsed DC vaccines without α-GalCer (DCV). DESIGN, SETTING AND PARTICIPANTS Single-centre blinded randomised controlled trial in patients ≥ 18 years old with histologically confirmed, fully resected stage II-IV malignant cutaneous melanoma, conducted between July 2015 and June 2018 at the Wellington Blood and Cancer Centre of the Capital and Coast District Health Board. INTERVENTIONS Stage I. Patients were randomised to two cycles of DCV or DCV + α-GalCer (intravenous dose of 10 × 106 cells, interval of 28 days). Stage II. Patients assigned to DCV + α-GalCer were randomised to two further cycles of DCV + α-GalCer or observation, while patients initially assigned to DCV crossed over to two cycles of DCV + α-GalCer. OUTCOME MEASURES Primary: Area under the curve (AUC) of mean NY-ESO-1-specific T cell count detected by ex vivo IFN-γ ELISpot in pre- and post-treatment blood samples, compared between treatment arms at Stage I. Secondary: Proportion of responders in each arm at Stage I; NKT cell count in each arm at Stage I; serum cytokine levels at Stage I; adverse events Stage I; T cell count for DCV + α-GalCer versus observation at Stage II, T cell count before versus after cross-over. RESULTS Thirty-eight patients gave written informed consent; 5 were excluded before randomisation due to progressive disease or incomplete leukapheresis, 17 were assigned to DCV, and 16 to DCV + α-GalCer. The vaccines were well tolerated and associated with increases in mean total T cell count, predominantly CD4+ T cells, but the difference between the treatment arms was not statistically significant (difference - 6.85, 95% confidence interval, - 21.65 to 7.92; P = 0.36). No significant improvements in T cell response were associated with DCV + α-GalCer with increased dosing, or in the cross-over. However, the NKT cell response to α-GalCer-loaded vaccines was limited compared to previous studies, with mean circulating NKT cell levels not significantly increased in the DCV + α-GalCer arm and no significant differences in cytokine response between the treatment arms. CONCLUSIONS A high population coverage of NY-ESO-1-specific T cell responses was achieved with a good safety profile, but we failed to demonstrate that loading with α-GalCer provided an additional advantage to the T cell response with this cellular vaccine design. CLINICAL TRIAL REGISTRATION ACTRN12612001101875. Funded by the Health Research Council of New Zealand.
Collapse
Affiliation(s)
- Nathaniel Dasyam
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Katrina J Sharples
- Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Catherine Barrow
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Ying Huang
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Evelyn Bauer
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Brigitta Mester
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Catherine E Wood
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Astrid Authier-Hall
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Marina Dzhelali
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Tess Ostapowicz
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Rajiv Kumar
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Jessica Lowe
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Alice Maxwell
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Olivia K Burn
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Geoffrey M Williams
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | - Sarah E Carley
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | | | - Jeremy Jones
- GlycoSyn, PO Box 31 310, Lower Hutt, 5040, New Zealand
| | - Susanna T S Chan
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Lower Hutt, 5046, New Zealand
| | - Victoria A Hinder
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Jerome Macapagal
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Monica McCusker
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
- Capital and Coast District Health Board, Private Bag 7902, Wellington, 6242, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Lower Hutt, 5046, New Zealand
| | - Michael P Findlay
- Cancer Trials New Zealand, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - P Rod Dunbar
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand
- School of Biological Sciences, University of Auckland, PO Box 92019, Auckland, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
215
|
Perez MC, Depalo DK, Zager JS. A safety review of recently approved and late-stage trial treatments for metastatic melanoma: systemic and regional therapies. Expert Opin Drug Saf 2023; 22:789-797. [PMID: 37551723 DOI: 10.1080/14740338.2023.2245333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION Advanced melanoma accounts for the majority of skin cancer-associated deaths. Over the past 15 years, there has been a dramatic change in the treatment options and prognosis for patients with advanced melanoma secondary to the development of novel systemic immunotherapies (IO) and targeted therapies. In addition to these novel systemic therapies, regional therapies (intralesional and perfusional) also continue to play a major role in the management of these patients. AREAS COVERED In this article, we review recent updates in the management of advanced melanoma via Medline (PubMed) and Google Scholar, including recently published trials in the metastatic, adjuvant, and neoadjuvant settings. We also review recently published trials for regional therapies and discuss future directions in the management of patients with advanced melanoma. EXPERT OPINION A significant portion of patients with advanced melanoma will develop recurrent or progressive disease following treatment with IO or targeted therapy. Therefore, identifying not only the appropriate therapeutic agent but also the sequence and duration of treatment is pivotal for these patients.
Collapse
Affiliation(s)
- Matthew C Perez
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Danielle K Depalo
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| |
Collapse
|
216
|
Ishizuki S, Nakamura Y. Role of Sentinel Lymph Node Biopsy for Skin Cancer Based on Clinical Studies. Cancers (Basel) 2023; 15:3291. [PMID: 37444401 DOI: 10.3390/cancers15133291] [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: 05/06/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The sentinel lymph node is the first lymph node from the primary tumor. Sentinel lymph node biopsy (SLNB) is a surgical procedure that can detect occult nodal metastasis with relatively low morbidity. It may also have a therapeutic effect via regional disease control. The Multicenter Selective Lymphadenectomy-I (MSLT-I) trial revealed a prognostic benefit from SLNB in melanoma patients. However, it remains unclear whether there is a prognostic benefit from SLNB in patients with nonmelanoma skin cancer owing to a lack of randomized prospective studies. Nevertheless, SLNB provides important information about nodal status, which is one of the strongest factors to predict prognosis and may guide additional nodal treatment. Currently, SLNB is widely used in the management of not only patients with melanoma but also those with nonmelanoma skin cancer. However, the utilization and outcomes of SLNB differ among skin cancers. In addition, SLNB is not recommended for routine use in all patients with skin cancer. In this review, we provide a summary of the role of SLNB and of the indications for SLNB in each skin cancer based on previously published articles.
Collapse
Affiliation(s)
- Shoichiro Ishizuki
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| |
Collapse
|
217
|
Fernandez MF, Choi J, Sosman J. New Approaches to Targeted Therapy in Melanoma. Cancers (Basel) 2023; 15:3224. [PMID: 37370834 PMCID: PMC10296143 DOI: 10.3390/cancers15123224] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
It was just slightly more than a decade ago when metastatic melanoma carried a dismal prognosis with few, if any, effective therapies. Since then, the evolution of cancer immunotherapy has led to new and effective treatment approaches for melanoma. However, despite these advances, a sizable portion of patients with advanced melanoma have de novo or acquired resistance to immune checkpoint inhibitors. At the same time, therapies (BRAF plus MEK inhibitors) targeting the BRAFV600 mutations found in 40-50% of cutaneous melanomas have also been critical for optimizing management and improving patient outcomes. Even though immunotherapy has been established as the initial therapy in most patients with cutaneous melanoma, subsequent effective therapy is limited to BRAFV600 melanoma. For all other melanoma patients, driver mutations have not been effectively targeted. Numerous efforts are underway to target melanomas with NRAS mutations, NF-1 LOF mutations, and other genetic alterations leading to activation of the MAP kinase pathway. In this era of personalized medicine, we will review the current genetic landscape, molecular classifications, emerging drug targets, and the potential for combination therapies for non-BRAFV600 melanoma.
Collapse
Affiliation(s)
| | | | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.F.F.); (J.C.)
| |
Collapse
|
218
|
Geoffrois L, Harlé A, Sahki N, Sikanja A, Granel-Brocard F, Hervieu A, Mortier L, Jeudy G, Michel C, Nardin C, Huin-Schohn C, Merlin JL. Personalized follow-up of circulating DNA in resected stage III/IV melanoma: PERCIMEL multicentric prospective study protocol. BMC Cancer 2023; 23:554. [PMID: 37328818 DOI: 10.1186/s12885-023-11029-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND With more than 15,000 new cases /year in France and 2,000 deaths, cutaneous melanoma represents approximately 4% of incidental cancers and 1.2% of cancer related deaths. In locally advanced (stage III) or resectable metastatic (stage IV) melanomas, medical adjuvant treatment is proposed and recent advances had shown the benefit of anti-PD1/PDL1 and anti-CTLA4 immunotherapy as well as anti-BRAF and anti-MEK targeted therapy in BRAF V600 mutated tumors. However, the recurence rate at one year is approximately 30% and justify extensive research of predictive biomarkers. If in metastatic disease, the follow-up of circulating tumor DNA (ctDNA) has been demonstrated, its interest in adjuvant setting remains to be precised, especially because of a lower detection rate. Further, the definition of a molecular response could prove useful to personalized treatment. METHODS PERCIMEL is an open prospective multicentric study executed through collaboration of the Institut de Cancérologie de Lorraine (non-profit comprehensive cancer center) and 6 French university and community hospitals. A total of 165 patients with resected stage III and IV melanoma, eligible to adjuvant imunotherapy or anti-BRAF/MEK kinase inhibitors will be included. The primary endpoint is the presence of ctDNA, 2 to 3 weeks after surgery, defined as mutated ctDNA copy number calculated as the allelic fraction of a clonal mutation relative to total ctDNA. Secondary endpoints are recurrence-free survival, distant metastasis-free survival and specific survival. We will follow ctDNA along treatment, quantitatively through ctDNA mutated copy number variation, qualitatively through the presence of cfDNA and its clonal evolution. Relative and absolute variations of ctDNA during follow-up will be also analyzed. PERCIMEL study aims at provide scientific evidence that ctDNA quantitative and qualitative variations can be used to predict the recurrence of patients with melanoma treated with adjuvant immunotherapy or kinase inhibitors, thus defining the notion of molecular recurrence.
Collapse
Affiliation(s)
- Lionnel Geoffrois
- Medical Oncology Department, Institut de Cancérologie de Lorraine, Vandoeuvre Les Nancy, France
| | - Alexandre Harlé
- Biopathology Department, Institut de Cancérologie de Lorraine, CNRS UMR7039 CRAN Université de Lorraine, Vandoeuvre Les Nancy, France
| | - Nassim Sahki
- Methodology Biostatistics Unit, Institut de Cancérologie de Lorraine, Vandoeuvre Les Nancy, France
| | - Aleksandra Sikanja
- Clinical Research Department, Institut de Cancérologie de Lorraine, Vandoeuvre Les Nancy, France
| | | | - Alice Hervieu
- Medical Oncology Department, Centre Georges François Leclerc, Dijon, France
| | - Laurent Mortier
- Dermatology Department CHRU Lille, Inserm U1189, Université de Lille, Lille, France
| | | | - Catherine Michel
- Dermatology Department, GHR Mulhouse Sud Alsace, Mulhouse, France
| | - Charlée Nardin
- Dermatology Department CHU Besançon, Inserm 1098 RIGHT Université Franche Comté, Besançon, France
| | - Cécile Huin-Schohn
- Clinical Research Department, Institut de Cancérologie de Lorraine, Vandoeuvre Les Nancy, France
| | - Jean-Louis Merlin
- Biopathology Department, Institut de Cancérologie de Lorraine, CNRS UMR7039 CRAN Université de Lorraine, Vandoeuvre Les Nancy, France.
- Clinical Research Department, Institut de Cancérologie de Lorraine, Vandoeuvre Les Nancy, France.
| |
Collapse
|
219
|
Żołnowska B, Sławiński J, Belka M, Bączek T, Chojnacki J, Kawiak A. Novel 2-alkythio-4-chloro- N-[imino(heteroaryl)methyl]benzenesulfonamide Derivatives: Synthesis, Molecular Structure, Anticancer Activity and Metabolic Stability. Int J Mol Sci 2023; 24:ijms24119768. [PMID: 37298719 DOI: 10.3390/ijms24119768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
A series of novel 2-alkythio-4-chloro-N-[imino-(heteroaryl)methyl]benzenesulfonamide derivatives, 8-24, were synthesized in the reaction of the N-(benzenesulfonyl)cyanamide potassium salts 1-7 with the appropriate mercaptoheterocycles. All the synthesized compounds were evaluated for their anticancer activity in HeLa, HCT-116 and MCF-7 cell lines. The most promising compounds, 11-13, molecular hybrids containing benzenesulfonamide and imidazole moieties, selectively showed a high cytotoxic effect in HeLa cancer cells (IC50: 6-7 μM) and exhibited about three times less cytotoxicity against the non-tumor cell line HaCaT cells (IC50: 18-20 μM). It was found that the anti-proliferative effects of 11, 12 and 13 were associated with their ability to induce apoptosis in HeLa cells. The compounds increased the early apoptotic population of cells, elevated the percentage of cells in the sub-G1 phase of the cell cycle and induced apoptosis through caspase activation in HeLa cells. For the most active compounds, susceptibility to undergo first-phase oxidation reactions in human liver microsomes was assessed. The results of the in vitro metabolic stability experiments indicated values of the factor t½ for 11-13 in the range of 9.1-20.3 min and suggested the hypothetical oxidation of these compounds to sulfenic and subsequently sulfinic acids as metabolites.
Collapse
Affiliation(s)
- Beata Żołnowska
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Jarosław Sławiński
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Jarosław Chojnacki
- Department of Inorganic Chemistry, Gdańsk University of Technology, ul. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Anna Kawiak
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, ul. Abrahama 58, 80-307 Gdańsk, Poland
| |
Collapse
|
220
|
O'Dwyer PJ, Gray RJ, Flaherty KT, Chen AP, Li S, Wang V, McShane LM, Patton DR, Tricoli JV, Williams PM, Iafrate AJ, Sklar J, Mitchell EP, Takebe N, Sims DJ, Coffey B, Fu T, Routbort M, Rubinstein LV, Little RF, Arteaga CL, Marinucci D, Hamilton SR, Conley BA, Harris LN, Doroshow JH. The NCI-MATCH trial: lessons for precision oncology. Nat Med 2023; 29:1349-1357. [PMID: 37322121 PMCID: PMC10612141 DOI: 10.1038/s41591-023-02379-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/28/2023] [Indexed: 06/17/2023]
Abstract
The NCI-MATCH (Molecular Analysis for Therapy Choice) trial ( NCT02465060 ) was launched in 2015 as a genomically driven, signal-seeking precision medicine platform trial-largely for patients with treatment-refractory, malignant solid tumors. Having completed in 2023, it remains one of the largest tumor-agnostic, precision oncology trials undertaken to date. Nearly 6,000 patients underwent screening and molecular testing, with a total of 1,593 patients (inclusive of continued accrual from standard next-generation sequencing) being assigned to one of 38 substudies. Each substudy was a phase 2 trial of a therapy matched to a genomic alteration, with a primary endpoint of objective tumor response by RECIST criteria. In this Perspective, we summarize the outcomes of the initial 27 substudies in NCI-MATCH, which met its signal-seeking objective with 7/27 positive substudies (25.9%). We discuss key aspects of the design and operational conduct of the trial, highlighting important lessons for future precision medicine studies.
Collapse
Affiliation(s)
| | - Robert J Gray
- Dana-Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | | | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Shuli Li
- Dana-Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | - Victoria Wang
- Dana-Farber Cancer Institute - ECOG-ACRIN Biostatistics Center, Boston, MA, USA
| | - Lisa M McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - David R Patton
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD, USA
| | - James V Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - P Mickey Williams
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - A John Iafrate
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | | | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - David J Sims
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brent Coffey
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD, USA
| | - Tony Fu
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Mark Routbort
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Larry V Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Richard F Little
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Carlos L Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | | | | | - Barbara A Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Lyndsay N Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
221
|
Moschella F, Buccione C, Ruspantini I, Castiello L, Rozo Gonzalez A, Iacobone F, Ferraresi V, Palermo B, Nisticò P, Belardelli F, Proietti E, Macchia I, Urbani F. Blood immune cells as potential biomarkers predicting relapse-free survival of stage III/IV resected melanoma patients treated with peptide-based vaccination and interferon-alpha. Front Oncol 2023; 13:1145667. [PMID: 37274275 PMCID: PMC10233106 DOI: 10.3389/fonc.2023.1145667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Despite the recent approval of several therapies in the adjuvant setting of melanoma, tumor relapse still occurs in a significant number of completely resected stage III-IV patients. In this context, the use of cancer vaccines is still relevant and may increase the response to immune checkpoint inhibitors. We previously demonstrated safety, immunogenicity and preliminary evidence of clinical efficacy in stage III/IV resected melanoma patients subjected to a combination therapy based on peptide vaccination together with intermittent low-dose interferon-α2b, with or without dacarbazine preconditioning (https://www.clinicaltrialsregister.eu/ctr-search/search, identifier: 2008-008211-26). In this setting, we then focused on pre-treatment patient immune status to highlight possible factors associated with clinical outcome. Methods Multiparametric flow cytometry was used to identify baseline immune profiles in patients' peripheral blood mononuclear cells and correlation with the patient clinical outcome. Receiver operating characteristic curve, Kaplan-Meier survival and principal component analyses were used to evaluate the predictive power of the identified markers. Results We identified 12 different circulating T and NK cell subsets with significant (p ≤ 0.05) differential baseline levels in patients who later relapsed with respect to patients who remained free of disease. All 12 parameters showed a good prognostic accuracy (AUC>0.7, p ≤ 0.05) and 11 of them significantly predicted the relapse-free survival. Remarkably, 3 classifiers also predicted the overall survival. Focusing on immune cell subsets that can be analyzed through simple surface staining, three subsets were identified, namely regulatory T cells, CD56dimCD16- NK cells and central memory γδ T cells. Each subset showed an AUC>0.8 and principal component analysis significantly grouped relapsing and non-relapsing patients (p=0.034). These three subsets were used to calculate a combination score that was able to perfectly distinguish relapsing and non-relapsing patients (AUC=1; p=0). Noticeably, patients with a combined score ≥2 demonstrated a strong advantage in both relapse-free (p=0.002) and overall (p=0.011) survival as compared to patients with a score <2. Discussion Predictive markers may be used to guide patient selection for personalized therapies and/or improve follow-up strategies. This study provides preliminary evidence on the identification of peripheral blood immune biomarkers potentially capable of predicting the clinical response to combined vaccine-based adjuvant therapies in melanoma.
Collapse
Affiliation(s)
- Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Buccione
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Andrea Rozo Gonzalez
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Floriana Iacobone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Virginia Ferraresi
- Department of Medical Oncology 1, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Filippo Belardelli
- Institute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
| | - Enrico Proietti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- Medical Biotechnology and Translational Medicine PhD School, II University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
222
|
Ziętek M, Teterycz P, Wierzbicki J, Jankowski M, Las-Jankowska M, Zegarski W, Piekarski J, Nejc D, Drucis K, Cybulska-Stopa B, Łobaziewicz W, Galwas K, Kamińska-Winciorek G, Zdzienicki M, Sryukina T, Ziobro A, Kluz A, Czarnecka AM, Rutkowski P. The Current Treatment Trends and Survival Patterns in Melanoma Patients with Positive Sentinel Lymph Node Biopsy (SLNB): A Multicenter Nationwide Study. Cancers (Basel) 2023; 15:2667. [PMID: 37345002 PMCID: PMC10216007 DOI: 10.3390/cancers15102667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND In melanoma treatment, an approach following positive sentinel lymph node biopsy (SLNB) has been recently deescalated from completion lymph node dissection (CLND) to active surveillance based on phase III trials data. In this study, we aim to evaluate treatment strategies in SLNB-positive melanoma patients in real-world practice. METHODS Five-hundred-fifty-seven melanoma SLNB-positive patients from seven comprehensive cancer centers treated between 2017 and 2021 were included. Kaplan-Meier methods and the Cox Proportional-Hazards Model were used for analysis. RESULTS The median follow-up was 25 months. Between 2017 and 2021, the percentage of patients undergoing CLND decreased (88-41%), while the use of adjuvant treatment increased (11-51%). The 3-year OS and RFS rates were 77.9% and 59.6%, respectively. Adjuvant therapy prolonged RFS (HR:0.69, p = 0.036)), but CLND did not (HR:1.22, p = 0.272). There were no statistically significant differences in OS for either adjuvant systemic treatment or CLND. Lower progression risk was also found, and time-dependent hazard ratios estimation in patients treated with systemic adjuvant therapy was confirmed (HR:0.20, p = 0.002 for BRAF inhibitors and HR:0.50, p = 0.015 for anti-PD-1 inhibitors). CONCLUSIONS Treatment of SLNB-positive melanoma patients is constantly evolving, and the role of surgery is currently rather limited. Whether CLND has been performed or not, in a group of SLNB-positive patients, adjuvant systemic treatment should be offered to all eligible patients.
Collapse
Affiliation(s)
- Marcin Ziętek
- Department of Oncology, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Surgical Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland;
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
- Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jędrzej Wierzbicki
- Department of Surgical Oncology, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland;
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Michał Jankowski
- Chair of Surgical Oncology, Collegium Medicum Nicolaus Copernicus University, Oncology Center—Prof Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (W.Z.)
| | - Manuela Las-Jankowska
- Chair of Surgical Oncology, Collegium Medicum Nicolaus Copernicus University, Oncology Center—Prof Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (W.Z.)
| | - Wojciech Zegarski
- Chair of Surgical Oncology, Collegium Medicum Nicolaus Copernicus University, Oncology Center—Prof Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (W.Z.)
| | - Janusz Piekarski
- Department of Surgical Oncology, Medical University of Lodz, 90-419 Lodz, Poland; (J.P.); (D.N.)
| | - Dariusz Nejc
- Department of Surgical Oncology, Medical University of Lodz, 90-419 Lodz, Poland; (J.P.); (D.N.)
- Nicolaus Copernicus Multidisciplinary Center for Oncology and Traumatology, 93-513 Lodz, Poland
| | - Kamil Drucis
- Department of Surgical Oncology, Gdansk Medical University, 80-210 Gdansk, Poland
| | - Bożena Cybulska-Stopa
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Cracow, Poland;
| | - Wojciech Łobaziewicz
- Department of Surgical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, 31-115 Cracow, Poland;
| | - Katarzyna Galwas
- 2nd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Grażyna Kamińska-Winciorek
- Department of Bone Marrow Transplantation and Onco-Hematology, Skin Cancer and Melanoma Team, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland;
| | - Marcin Zdzienicki
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
| | - Tatsiana Sryukina
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Ziobro
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Agnieszka Kluz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Centers, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland (M.Z.); (A.Z.); (A.M.C.); (P.R.)
| |
Collapse
|
223
|
Lucas MW, Versluis JM, Rozeman EA, Blank CU. Personalizing neoadjuvant immune-checkpoint inhibition in patients with melanoma. Nat Rev Clin Oncol 2023; 20:408-422. [PMID: 37147419 DOI: 10.1038/s41571-023-00760-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 05/07/2023]
Abstract
Neoadjuvant immune-checkpoint inhibition is a promising emerging treatment approach for patients with surgically resectable macroscopic stage III melanoma. The neoadjuvant setting provides an ideal platform for personalized therapy owing to the very homogeneous nature of the patient population and the opportunity for pathological response assessments within several weeks of starting treatment, thereby facilitating the efficient identification of novel biomarkers. A pathological response to immune-checkpoint inhibitors has been shown to be a strong surrogate marker of both recurrence-free survival and overall survival, enabling timely analyses of the efficacy of novel therapies in patients with early stage disease. Patients with a major pathological response (defined as the presence of ≤10% viable tumour cells) have a very low risk of recurrence, which offers an opportunity to adjust the extent of surgery and any subsequent adjuvant therapy and follow-up monitoring. Conversely, patients who have only a partial pathological response or who do not respond to neoadjuvant therapy still might benefit from therapy escalation and/or class switch during adjuvant therapy. In this Review, we outline the concept of a fully personalized neoadjuvant treatment approach exemplified by the current developments in neoadjuvant therapy for patients with resectable melanoma, which could provide a template for the development of similar approaches for patients with other immune-responsive cancers in the near future.
Collapse
Affiliation(s)
- Minke W Lucas
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Judith M Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Elisa A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
224
|
Gouda MA, Subbiah V. Expanding the Benefit: Dabrafenib/Trametinib as Tissue-Agnostic Therapy for BRAF V600E-Positive Adult and Pediatric Solid Tumors. Am Soc Clin Oncol Educ Book 2023; 43:e404770. [PMID: 37159870 DOI: 10.1200/edbk_404770] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The recent US Food and Drug Administration (FDA) approval of the dabrafenib/trametinib combination as a tissue-agnostic treatment for solid tumors with BRAF V600E mutation is the result of more than 20 years of extensive research into BRAF mutations in human cancer, the underlying biological mechanisms that drive BRAF-mediated tumor growth, and the clinical testing and refinement of selective RAF and MEK kinase inhibitors. Such approval marks a significant achievement in the field of oncology and represents a major step forward in our ability to treat cancer. Early evidence supported the use of dabrafenib/trametinib combination in melanoma, non-small-cell lung cancer, and anaplastic thyroid cancer. Furthermore, data from basket trials have demonstrated consistently good response rates in various tumors, including biliary tract cancer, low-grade glioma, high-grade glioma, hairy cell leukemia, and multiple other malignancies, which has been the basis for FDA approval of a tissue-agnostic indication in adult and pediatric patients with BRAF V600E-positive solid tumors. From a clinical standpoint, our review delves into the efficacy of the dabrafenib/trametinib combination for BRAF V600E-positive tumors: examining the underlying rationale for its use, evaluating the latest evidence on its potential benefits, and discussing the possible associated adverse effects and strategies to minimize their impact. Additionally, we explore potential resistance mechanisms and future landscape of BRAF-targeted therapies.
Collapse
Affiliation(s)
- Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Clinical Oncology, Faculty of Medicine, Menoufia University, Shebin Al-Kom, Egypt
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX
- MD Anderson Cancer Network, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
225
|
Broman KK, Hughes TM, Bredbeck BC, Sun J, Kirichenko D, Carr MJ, Sharma A, Bartlett EK, Nijhuis AAG, Thompson JF, Hieken TJ, Kottschade L, Downs J, Gyorki DE, Stahlie E, van Akkooi A, Ollila DW, O'shea K, Song Y, Karakousis G, Moncrieff M, Nobes J, Vetto J, Han D, Hotz M, Farma JM, Deneve JL, Fleming MD, Perez M, Baecher K, Lowe M, Bagge RO, Mattsson J, Lee AY, Berman RS, Chai H, Kroon HM, Teras J, Teras RM, Farrow NE, Beasley GM, Hui JYC, Been L, Kruijff S, Sinco B, Sarnaik AA, Sondak VK, Zager JS, Dossett LA. International Center-Level Variation in Utilization of Completion Lymph Node Dissection and Adjuvant Systemic Therapy for Sentinel Lymph Node-Positive Melanoma at Major Referral Centers. Ann Surg 2023; 277:e1106-e1115. [PMID: 35129464 PMCID: PMC10097464 DOI: 10.1097/sla.0000000000005370] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of this study was to determine overall trends and center-level variation in utilization of completion lymph node dissection (CLND) and adjuvant systemic therapy for sentinel lymph node (SLN)-positive melanoma. SUMMARY BACKGROUND DATA Based on recent clinical trials, management options for SLN-positive melanoma now include effective adjuvant systemic therapy and nodal observation instead of CLND. It is unknown how these findings have shaped practice or how these contemporaneous developments have influenced their respective utilization. METHODS We performed an international cohort study at 21 melanoma referral centers in Australia, Europe, and the United States that treated adults with SLN-positive melanoma and negative distant staging from July 2017 to June 2019. We used generalized linear and multinomial logistic regression models with random intercepts for each center to assess center-level variation in CLND and adjuvant systemic treatment, adjusting for patient and disease-specific characteristics. RESULTS Among 1109 patients, performance of CLND decreased from 28% to 8% and adjuvant systemic therapy use increased from 29 to 60%. For both CLND and adjuvant systemic treatment, the most influential factors were nodal tumor size, stage, and location of treating center. There was notable variation among treating centers in management of stage IIIA patients and use of CLND with adjuvant systemic therapy versus nodal observation alone for similar risk patients. CONCLUSIONS There has been an overall decline in CLND and simultaneous adoption of adjuvant systemic therapy for patients with SLN-positive melanoma though wide variation in practice remains. Accounting for differences in patient mix, location of care contributed significantly to the observed variation.
Collapse
Affiliation(s)
- Kristy K Broman
- Moffitt Cancer Center, Tampa, FL
- University of South Florida Morsani College of Medicine, Tampa, FL
- University of Alabama at Birmingham, Birmingham, AL
| | | | | | | | | | | | | | | | - Amanda A G Nijhuis
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | - Emma Stahlie
- Netherlands Cancer institute, Amsterdam, The Netherlands
| | | | | | | | - Yun Song
- University of Gothenburg, Gothenburg, Sweden
| | | | - Marc Moncrieff
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Jenny Nobes
- Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - John Vetto
- Oregon Health & Science University, Portland, OR
| | - Dale Han
- Oregon Health & Science University, Portland, OR
| | | | | | | | | | | | | | | | | | - Jan Mattsson
- University Medical Center, Groningen, Netherlands
| | | | | | - Harvey Chai
- Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Hidde M Kroon
- Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Juri Teras
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Roland M Teras
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | | | | | | | | | | | | | - Amod A Sarnaik
- Moffitt Cancer Center, Tampa, FL
- University of South Florida Morsani College of Medicine, Tampa, FL
| | - Vernon K Sondak
- Moffitt Cancer Center, Tampa, FL
- University of South Florida Morsani College of Medicine, Tampa, FL
| | - Jonathan S Zager
- Moffitt Cancer Center, Tampa, FL
- University of South Florida Morsani College of Medicine, Tampa, FL
| | | |
Collapse
|
226
|
Yang TT, Yu S, Ke CLK, Cheng ST. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes (Basel) 2023; 14:genes14051021. [PMID: 37239381 DOI: 10.3390/genes14051021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/25/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Melanoma is one of the most aggressive malignancies of the skin. The genetic composition of melanoma is complex and varies among different subtypes. With the aid of recent technologies such as next generation sequencing and single-cell sequencing, our understanding of the genomic landscape of melanoma and its tumor microenvironment has become increasingly clear. These advances may provide explanation to the heterogenic treatment outcomes of melanoma patients under current therapeutic guidelines and provide further insights to the development of potential new therapeutic targets. Here, we provide a comprehensive review on the genetics related to melanoma tumorigenesis, metastasis, and prognosis. We also review the genetics affecting the melanoma tumor microenvironment and its relation to tumor progression and treatment.
Collapse
Affiliation(s)
- Ting-Ting Yang
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, Pingtung Hospital, Ministry of Health and Welfare, Pingtung 900, Taiwan
| | - Sebastian Yu
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chiao-Li Khale Ke
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Psychiatry, Kaohsiung Municipal SiaoGang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Shih-Tsung Cheng
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
227
|
Long GV, Menzies AM, Scolyer RA. Neoadjuvant Checkpoint Immunotherapy and Melanoma: The Time Is Now. J Clin Oncol 2023:JCO2202575. [PMID: 37104746 DOI: 10.1200/jco.22.02575] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
The role of neoadjuvant therapy is undergoing an historic shift in oncology. The emergence of potent immunostimulatory anticancer agents has transformed neoadjuvant therapy from a useful tool in minimizing surgical morbidity to a life-saving treatment with curative promise, led by research in the field of melanoma. Health practitioners have witnessed remarkable improvements in melanoma survival outcomes over the past decade, beginning with checkpoint immunotherapies and BRAF-targeted therapies in the advanced setting that were successfully adopted into the postsurgical adjuvant setting for high-risk resectable disease. Despite substantial reductions in postsurgical recurrence, high-risk resectable melanoma has remained a life-altering and potentially fatal disease. In recent years, data from preclinical models and early-phase clinical trials have pointed to the potential for greater clinical efficacy when checkpoint inhibitors are administered in the neoadjuvant rather than adjuvant setting. Early feasibility studies showed impressive pathologic response rates to neoadjuvant immunotherapy, which were associated with recurrence-free survival rates of over 90%. Recently, the randomized phase II SWOG S1801 trial (ClinicalTrials.gov identifier: NCT03698019) reported a 42% reduction in 2-year event-free survival risk with neoadjuvant versus adjuvant pembrolizumab in resectable stage IIIB-D/IV melanoma (72% v 49%; hazard ratio, 0.58; P = .004), establishing neoadjuvant single-agent immunotherapy as a new standard of care. A randomized phase III trial of neoadjuvant immunotherapy in resectable stage IIIB-D melanoma, NADINA (ClinicalTrials.gov identifier: NCT04949113), is ongoing, as are feasibility studies in high-risk stage II disease. With a swathe of clinical, quality-of-life, and economic benefits, neoadjuvant immunotherapy has the potential to redefine the contemporary management of resectable tumors.
Collapse
Affiliation(s)
- 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
- Royal North Shore Hospital, Sydney, NSW, Australia
- Mater Hospital, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore Hospital, Sydney, NSW, Australia
- Mater Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- 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 Prince Alfred Hospital, Sydney, NSW, Australia
- NSW Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
228
|
Kinaschuk K, Cheng T, Brenn T, McKinnon JG, Temple-Oberle C. Not Waiting to Progress; How the COVID-19 Pandemic Nudged Neoadjuvant Therapy for Stage III Locally Advanced Melanoma Patients. Curr Oncol 2023; 30:4402-4411. [PMID: 37232793 DOI: 10.3390/curroncol30050335] [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: 01/24/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 05/27/2023] Open
Abstract
Background: Early-phase neoadjuvant trials have demonstrated promising results in the utility of upfront immunotherapy in locally advanced stage III melanoma and unresected nodal disease. Secondary to these results and the COVID-19 pandemic, this patient population, traditionally managed through surgical resection and adjuvant immunotherapy, received a novel treatment strategy of neoadjuvant therapy (NAT). Methods: Patients with node-positive disease, who faced surgical delays secondary to COVID-19, were treated with NAT, followed by surgery. Demographic, tumour, treatment and response data were collected through a retrospective chart review. Biopsy specimens were analysed prior to the initiation of NAT, and therapy response was analysed following surgical resection. NAT tolerability was recorded. Results: Six patients were included in this case series; four were treated with nivolumab alone, one with ipilimumab and nivolumab and one with dabrafenib and trametinib. Twenty-two incidents of adverse events were reported, with the majority (90.9%) being classified as grade one or two. All patients underwent surgical resection: three out of six patients following two NAT cycles, two following three cycles and one following six cycles. Surgically resected samples were histopathologically evaluated for the presence of disease. Five out of six patients (83%) had ≤1 positive lymph node. One patient showed extracapsular extension. Four patients demonstrated complete pathological response; two had persisting viable tumour cells. Conclusions: In this case series, we outlined how in response to surgical delays secondary to the COVID-19 pandemic, NAT was successfully applied to achieve promising treatment response in patients with locally advanced stage III melanoma.
Collapse
Affiliation(s)
- Katie Kinaschuk
- Tom Baker Cancer Centre, 1331-29 Street NW, Calgary, AB T2N 4N2, Canada
| | - Tina Cheng
- Tom Baker Cancer Centre, 1331-29 Street NW, Calgary, AB T2N 4N2, Canada
| | - Thomas Brenn
- Tom Baker Cancer Centre, 1331-29 Street NW, Calgary, AB T2N 4N2, Canada
| | | | | |
Collapse
|
229
|
Ascierto PA, Blank C, Eggermont AM, Garbe C, Gershenwald JE, Hamid O, Hauschild A, Luke JJ, Mehnert JM, Sosman JA, Tawbi HA, Mandalà M, Testori A, Caracò C, Osman I, Puzanov I. The "Great Debate" at Melanoma Bridge 2022, Naples, December 1st-3rd, 2022. J Transl Med 2023; 21:265. [PMID: 37072748 PMCID: PMC10114457 DOI: 10.1186/s12967-023-04100-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
The Great Debate session at the 2022 Melanoma Bridge congress (December 1-3) featured counterpoint views from leading experts on five contemporary topics of debate in the management of melanoma. The debates considered the choice of anti-lymphocyte-activation gene (LAG)-3 therapy or ipilimumab in combination with anti-programmed death (PD)-1 therapy, whether anti-PD-1 monotherapy is still acceptable as a comparator arm in clinical trials, whether adjuvant treatment of melanoma is still a useful treatment option, the role of adjuvant therapy in stage II melanoma, what role surgery will continue to have in the treatment of melanoma. As is customary in the Melanoma Bridge Great Debates, the speakers are invited by the meeting Chairs to express one side of the assigned debate and the opinions given may not fully reflect personal views. Audiences voted in favour of either side of the argument both before and after each debate.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | | | - Alexander M Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, The Netherlands
- Comprehensive Cancer Center München, Technical University München & Ludwig Maximiliaan University, München, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, CA, USA
| | - Axel Hauschild
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Jason J Luke
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, USA
| | - Janice M Mehnert
- Perlmutter Cancer Center of NYU Langone/NYU Grossman School of Medicine, New York, NY, USA
| | - Jeffrey A Sosman
- Robert H Lurie Comprehensive Cancer Center, Northwestern University Medical Center, Chicago, IL, USA
| | - Hussein A Tawbi
- MD Anderson Brain Metastasis Clinic UT, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Alessandro Testori
- Image regenerative clinic Milan, Italy; EORTC Melanoma Group, Brussels, Belgium
| | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori "Fondazione Pascale" IRCCS, Naples, Italy
| | - Iman Osman
- Rudolf L. Baer, NYU Langone Medical Center, New York, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
230
|
Pérez-Morales J, Broman KK, Bettampadi D, Haver MK, Zager JS, Schabath MB. Recurrence Patterns for Regionally Metastatic Melanoma Treated in the Era of Adjuvant Therapy: A Systematic Review and Meta-Analysis. Ann Surg Oncol 2023; 30:2364-2374. [PMID: 36479663 DOI: 10.1245/s10434-022-12866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The purpose of this systematic review was to examine the timing and patterns of recurrence for patients with regionally metastatic melanoma on the basis of nodal management and receipt of adjuvant therapy. METHODS We identified randomized controlled trials and non-randomized studies published between 2010 and 2020 that reported timing and/or patterns of recurrence. We evaluated recurrence-free survival (RFS), location of recurrence, and surveillance strategy on the basis of receipt of adjuvant systemic therapy and nodal management with observation versus completion dissection. We compared differences in patterns of recurrence across studies using RevMan. RFS was evaluated graphically using point estimates and confidence intervals. RESULTS Among the 19 publications, there was wide variation in study populations, imaging surveillance regimens, and format of recurrence reporting. Patterns of disease recurrence did not differ between adjuvant and placebo/observation groups. A total of 11 studies reported RFS at variable time intervals, which ranged in adjuvant therapy groups (38-88% at 1 year, 29-67% at 2 years, 33-58% at 3 years, and 34-53% at 5 years) and placebo/observation groups (47-63% at 1 year, 39-47% at 2 years, 33-68% at 3 years, and 57% at 5 years). Anti-PD-1 immune therapy and BRAF/MEK inhibitor therapy were superior to placebo at year 1. DISCUSSION We found that adjuvant treatment improved RFS but did not alter the patterns of disease recurrence compared with patients managed without adjuvant systemic treatment. Future studies should separately report sites of disease recurrence on the basis of specific adjuvant systemic treatment and surveillance practices to better advise patients about their patterns and risk of recurrence.
Collapse
Affiliation(s)
- Jaileene Pérez-Morales
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| | - Kristy K Broman
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Deepti Bettampadi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Mary Katherine Haver
- Moffitt Biomedical Library, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
231
|
Schoenfeldt T, Thompson JF, Lo S, Drzewiecki KT, Stretch J, Saw RPM, Spillane A, Shannon K, Uren RF, Chakera AH, Nieweg OE. Prognostic Significance and Management of Sentinel Nodes in the Triangular Intermuscular Space of Patients with Melanoma. Ann Surg Oncol 2023; 30:2354-2361. [PMID: 36463358 DOI: 10.1245/s10434-022-12840-2] [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: 08/15/2022] [Accepted: 11/03/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND The clinical significance of sentinel nodes (SNs) in the triangular intermuscular space (TIS) of patients with melanoma is poorly understood. This study aimed to determine their incidence and positivity rate, and to report their management and patient outcomes. METHODS This was a single-institution retrospective cohort study of patients with unilateral or bilateral TIS SNs on lymphoscintigraphy treated between 1992 and 2017. Recurrence-free survival was analyzed. RESULTS Lymphoscintigraphy identified TIS SNs in 266 patients. They were bilateral in 17 patients. Of the 2296 patients with a melanoma on the upper back, 259 (11%) had TIS SNs. Procurement of SNs was not attempted in 122 (43%) of the 283 cases and failed in 11 cases (7%). An SN was successfully retrieved from the TIS in 145 patients (53%) and contained metastasis in 18 of 150 TIS SNs. This was the only positive SN in 12 patients (8%), upstaging all of them. Of the 18 patients with a positive SN in the TIS, 9 (50%) underwent completion axillary lymph node dissection, but no additional involved nodes were found in any of these patients. Recurrence in the TIS was observed in six patients (5%), none of whom had their TIS SN surgically pursued previously. CONCLUSIONS Lymphoscintigraphy showed TIS SNs in 11% of patients with melanomas on their upper back. In such cases, retrieval of TIS SNs is required for accurate staging and to minimize the risk of TIS recurrence.
Collapse
Affiliation(s)
- Trine Schoenfeldt
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia.
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| | - Serigne Lo
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
| | - Krzysztof T Drzewiecki
- Department of Plastic Surgery, Breast Surgery and Burns, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, The University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Stretch
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Andrew Spillane
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Breast and Melanoma Surgery Unit, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - Kerwin Shannon
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Roger F Uren
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Alfred Nuclear Medicine and Ultrasound, Sydney, NSW, Australia
| | - Annette H Chakera
- Department of Plastic Surgery, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Omgo E Nieweg
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Road, North Sydney, NSW, 2060, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
232
|
Chen DY, Sutton LA, Ramakrishnan SM, Duncavage EJ, Heath SE, Compton LA, Miller CA, Ley TJ. Melanoma in a patient with DNMT3A overgrowth syndrome. Cold Spring Harb Mol Case Stud 2023; 9:a006267. [PMID: 37160317 PMCID: PMC10240841 DOI: 10.1101/mcs.a006267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 05/11/2023] Open
Abstract
Alterations in epigenetic regulators are increasingly recognized as early events in tumorigenesis; thus, patients with acquired or inherited variants in epigenetic regulators may be at increased risk for developing multiple types of cancer. DNMT3A overgrowth syndrome (DOS), caused by germline pathogenic variants in the DNA methyltransferase gene DNMT3A, has been associated with a predisposition toward development of hematopoietic and neuronal malignancies. DNMT3A deficiency has been described to promote keratinocyte proliferation in mice. Although altered DNA methylation patterns are well-recognized in melanoma, the role of DNA methyltransferases in melanoma pathogenesis is not clear. We report the case of an adult DOS patient with a germline DNMT3A loss-of-function mutation, who developed an early-onset melanoma with regional lymph node metastatic disease. Exome sequencing of the primary tumor identified an additional acquired, missense DNMT3A mutation in the dominant tumor clone, suggesting that the loss of DNMT3A function was relevant for the development of this tumor.
Collapse
Affiliation(s)
- David Y Chen
- Division of Dermatology, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Leslie A Sutton
- Division of Dermatology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Sai Mukund Ramakrishnan
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Sharon E Heath
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Leigh A Compton
- Division of Dermatology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Christopher A Miller
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Timothy J Ley
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
233
|
Weber JS, Poretta T, Stwalley BD, Sakkal LA, Du EX, Wang T, Chen Y, Wang Y, Betts KA, Shoushtari AN. Nivolumab versus placebo as adjuvant therapy for resected stage III melanoma: a propensity weighted indirect treatment comparison and number needed to treat analysis for recurrence-free survival and overall survival. Cancer Immunol Immunother 2023; 72:945-954. [PMID: 36197494 PMCID: PMC10025222 DOI: 10.1007/s00262-022-03302-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/23/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Recurrence-free survival (RFS) and overall survival (OS) data for adjuvant nivolumab versus placebo (proxy for routine surveillance) in patients with high-risk, resected melanoma are lacking. This post hoc, indirect treatment comparison (ITC) used pooled data from the phase 3 EORTC 18,071 (ipilimumab vs. placebo) and CheckMate 238 (nivolumab vs. ipilimumab) trials to assess RFS and OS with nivolumab versus placebo and the numbers needed to treat (NNT) over 4 years. METHODS Patients with resected stage IIIB-C cutaneous melanoma (American Joint Committee on Cancer seventh edition) were included. Inverse probability treatment weighting (IPTW) was used to balance baseline characteristics. RFS NNTs were calculated for nivolumab versus ipilimumab and placebo. OS NNTs were calculated for nivolumab versus placebo. To adjust for different post-recurrence treatments, the difference in post-recurrence survival between the two ipilimumab arms was added to OS of the placebo arm. RESULTS This ITC included 278, 643, and 365 patients treated with nivolumab, ipilimumab, and placebo, respectively. Following IPTW, nivolumab was associated with improved RFS versus placebo (hazard ratio [HR]: 0.49; 95% confidence interval [CI] 0.39-0.61) and ipilimumab (HR: 0.69; 95% CI 0.56-0.85). RFS NNT was 4.2 for nivolumab versus placebo and 8.9 for nivolumab versus ipilimumab. After post-recurrence survival adjustment, weighted 4-year OS rates were 75.8% for nivolumab and 64.1% for placebo; OS NNT for nivolumab versus placebo was 8.5. CONCLUSIONS In patients with resected stage IIIB-C cutaneous melanoma in this ITC, nivolumab improved RFS versus placebo and ipilimumab, and OS versus placebo after post-recurrence survival adjustment.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY, USA.
| | | | | | | | - Ella X Du
- Analysis Group, Inc., Los Angeles, CA, USA
| | | | - Yan Chen
- Analysis Group, Inc., Los Angeles, CA, USA
| | - Yan Wang
- Analysis Group, Inc., Los Angeles, CA, USA
| | | | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
234
|
Li Z, Zhou H, Xia Z, Xia T, Du G, Franziska SD, Li X, Zhai X, Jin B. HMGA1 augments palbociclib efficacy via PI3K/mTOR signaling in intrahepatic cholangiocarcinoma. Biomark Res 2023; 11:33. [PMID: 36978140 PMCID: PMC10053751 DOI: 10.1186/s40364-023-00473-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (iCCA) is a highly aggressive cancer that is challenging to diagnose at an early stage. Despite recent advances in combination chemotherapy, drug resistance limits the therapeutic value of this regimen. iCCA reportedly harbors high HMGA1 expression and pathway alterations, especially hyperactivation of the CCND1/CDK4/CDK6 and PI3K signaling pathway. In this study, we explored the potential of targeting CDK4/6 and PI3K inhibition to treat iCCA. METHODS The significance of HMGA1 in iCCA was investigated with in vitro/vivo experiments. Western blot, qPCR, dual-luciferase reporter and immunofluorescence assays were performed to examine the mechanism of HMGA1 induced CCND1 expression. CCK-8, western blot, transwell, 3D sphere formation and colony formation assays were conducted to predict the potential role of CDK4/6 inhibitors PI3K/mTOR inhibitors in iCCA treatment. Xenograft mouse models were also used to determine the efficacy of combination treatment strategies related to HMGA1 in iCCA. RESULTS HMGA1 promoted the proliferation, epithelial-mesenchymaltransition (EMT), metastasis and stemness of iCCA. In vitro studies showed that HMGA1 induced CCND1 expression via promoting CCND1 transcription and activating the PI3K signaling pathway. Palbociclib(CDK4/6 inhibitor) could suppress iCCA proliferation, migration and invasion, especially during the first 3 days. Although there was more stable attenuation of growth in the HIBEpic model, we observed substantial outgrowth in each hepatobiliary cancer cell model. PF-04691502(PI3K/mTOR inhibitor) exhibited similar effects to palbociclib. Compared with monotherapy, the combination retained effective inhibition for iCCA through the more potent and steady inhibition of CCND1, CDK4/6 and PI3K pathway. Furthermore, more significant inhibition of the common downstream signaling pathways is observed with the combination compared to monotherapy. CONCLUSIONS Our study reveals the potential therapeutic role of dual inhibition of CDK4/6 and PI3K/mTOR pathways in iCCA, and proposes a new paradigm for the clinical treatment of iCCA.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Huaxin Zhou
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
- The Second Clinical College of Shandong University, Jinan, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tong Xia
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
| | - Gang Du
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
| | - Strohmer Dorothee Franziska
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Xiaoming Li
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| | - Bin Jin
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
235
|
Robak T, Robak P. Refractory and relapsed hairy-cell leukemia (HCL): casting light on promising experimental drugs in clinical trials. Expert Opin Investig Drugs 2023; 32:311-324. [PMID: 36931901 DOI: 10.1080/13543784.2023.2193323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
INTRODUCTION Hairy cell leukemia (HCL) is a rare subtype of indolent lymphoid leukemia originating from a mature B lymphocyte. The standard first-line treatment for classic HCL, and HCL variant (HCLv), consists of purine nucleoside analogs (PNA), with or without rituximab. However, almost half of patients relapse and require subsequent therapy. AREAS COVERED This article summarizes recent achievements in the treatment of relapsed and refractory HCL. A literature search was conducted of the PubMed and MEDLINE database for articles in English. Publications from 2010 through January 2023 were scrutinized. The search terms used were hairy cell leukemia in conjunction with BRAF inhibitors, Bruton's tyrosine kinase (BTK) inhibitors, CD20 monoclonal antibodies, relapsed, refractory and variant.The growing understanding of HCL biology has allowed the design of several new, chemotherapy-free targeted drugs which have demonstrated encouraging efficacy in early clinical trials. EXPERT OPINION Novel drugs will soon be available to assist standard therapy for HCL and HCLv among patients with suboptimal results following PNA treatment. In particular, the BRAF inhibitors vemurafenib and dabrafenib, with or without rituximab, have revolutionized treatment of patients with relapsed or refractory disease.
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Łódź, Poland
- Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| | - Paweł Robak
- Department of Experimental Hematology, Medical University of Lodz, Lodz, Poland
- Department of Hematooncology, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
236
|
Senders ZJ, Bartlett EK, Mouw TJ, McMasters KM, Egger ME. Does Stage Migration Occur as a Consequence of Omitting Completion Lymph Node Dissection for Melanoma? Ann Surg Oncol 2023; 30:3648-3654. [PMID: 36934378 DOI: 10.1245/s10434-023-13342-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 02/19/2023] [Indexed: 03/20/2023]
Abstract
INTRODUCTION Completion lymph node dissection (CLND) is no longer recommended routinely in the treatment of melanoma. CLND omission may understage patients for whom the distinction between stage IIIA and IIIB-C could alter adjuvant therapy recommendations. The aim of this study is to determine if stage migration has occurred with the declining use of CLND. METHODS Patients with clinically node-negative ≥ T1b cutaneous melanoma were identified from the National Cancer Database (NCDB) from 2012 to 2018. CLND utilization and changes in AJCC staging were analyzed. Patients undergoing sentinel lymph node biopsy (SLNB) alone were compared with those undergoing SLNB + CLND. RESULTS Overall, 68,933 patients met inclusion criteria and 60,536 underwent SLNB, of which 9031 (14.9%) were tumor positive. CLND was performed in 3776 (41.8%). Patients undergoing CLND were younger (58 versus 62 years, p < 0.0001) and more likely male (61.5% versus 57.9%, p = 0.0005). Patients were more likely to have an N classification >N1a if they received SLNB + CLND (36.8%) versus SLNB alone (19.3%), p < 0.0001. This translated to a small difference in stage IIIA patients between groups (SLNB alone 34.0%, SLNB + CLND 31.8%, p < 0.0001). Of the patients with T1b/T2a tumors who would be upstaged from IIIA to IIIC with identification of additional positive nodes, IIIC incidence was only slightly higher after SLNB + CLND compared with SLNB alone (4.4% versus 1.1%, p < 0.0001). CLND utilization dramatically decreased from 59% in 2012 to 12.6% in 2018, p < 0.0001. However, the incidence of stage IIIA disease for all patients remained stable over the 7-year study period. CONCLUSIONS While the utilization of CLND after positive SLNB has declined dramatically in the last 7 years, stage migration that may affect adjuvant therapy decisions has not occurred to a clinically meaningful degree.
Collapse
Affiliation(s)
- Zachary J Senders
- Division of Surgical Oncology, Hiram C. Polk, Jr, MD Department of Surgery, University of Louisville, Louisville, KY, USA.
| | - Edmund K Bartlett
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tyler J Mouw
- Division of Surgical Oncology, Hiram C. Polk, Jr, MD Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Kelly M McMasters
- Division of Surgical Oncology, Hiram C. Polk, Jr, MD Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Michael E Egger
- Division of Surgical Oncology, Hiram C. Polk, Jr, MD Department of Surgery, University of Louisville, Louisville, KY, USA
| |
Collapse
|
237
|
Bialves TS, Bastos Junior CLQ, Cordeiro MF, Boyle RT. Snake venom, a potential treatment for melanoma. A systematic review. Int J Biol Macromol 2023; 231:123367. [PMID: 36690229 DOI: 10.1016/j.ijbiomac.2023.123367] [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: 10/11/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Despite advances in treating patients with melanoma, there are still many treatment challenges to overcome. Studies with snake venom-derived proteins/peptides describe their binding potential, and inhibition of some proliferative mechanisms in melanoma. The combined use of these compounds with current therapies could be the strategic gap that will help us discover more effective treatments for melanoma. The present study aimed to carry out a systematic review identifying snake venom proteins and peptides described in the literature with antitumor, antimetastatic, or antiangiogenic effects on melanoma and determine the mechanisms of action that lead to these anti-tumor effects. Snake venoms contain proteins and peptides which are antiaggregant, antimetastatic, and antiangiogenic. The in vivo results are encouraging, considering the reduction of metastases and tumor size after treatment. In addition to these results, it was reported that these venom compounds could act in combination with chemotherapeutics (Acurhagin-C; Macrovipecetin), sensitizing and preparing tumor cells for treatment. There is a consensus that snake venom is a promising strategy for the improvement of antimelanoma therapies, but it has been little explored in the current context, combined with inhibitors, immunotherapy or tumor microenvironment, for example. We suggest Lebein as a candidate for combination therapy with BRAF inhibitors.
Collapse
Affiliation(s)
- Tatiane Senna Bialves
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil.
| | - Claudio L Q Bastos Junior
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil
| | - Marcos Freitas Cordeiro
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Universidade do Oeste de Santa Catarina - UNOESC, Rua Roberto Trompovski 224, Joaçaba, Santa Catarina, CEP 89600-000, Brazil.
| | - Robert Tew Boyle
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil; Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Rio Grande do Sul 96203-900, Brazil
| |
Collapse
|
238
|
Viganò M, La Milia M, Grassini MV, Pugliese N, De Giorgio M, Fagiuoli S. Hepatotoxicity of Small Molecule Protein Kinase Inhibitors for Cancer. Cancers (Basel) 2023; 15:cancers15061766. [PMID: 36980652 PMCID: PMC10046041 DOI: 10.3390/cancers15061766] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Small molecule protein kinase inhibitors (PKIs) have become an effective strategy for cancer patients. However, hepatotoxicity is a major safety concern of these drugs, since the majority are reported to increase transaminases, and few of them (Idelalisib, Lapatinib, Pazopanib, Pexidartinib, Ponatinib, Regorafenib, Sunitinib) have a boxed label warning. The exact rate of PKI-induced hepatoxicity is not well defined due to the fact that the majority of data arise from pre-registration or registration trials on fairly selected patients, and the post-marketing data are often based only on the most severe described cases, whereas most real practice studies do not include drug-related hepatotoxicity as an end point. Although these side effects are usually reversible by dose adjustment or therapy suspension, or by switching to an alternative PKI, and fatality is uncommon, all patients undergoing PKIs should be carefully pre-evaluated and monitored. The management of this complication requires an individually tailored reappraisal of the risk/benefit ratio, especially in patients who are responding to therapy. This review reports the currently available data on the risk and management of hepatotoxicity of all the approved PKIs.
Collapse
Affiliation(s)
- Mauro Viganò
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Correspondence: ; Tel.: +39-035-2674259; Fax: +39-035-2674964
| | - Marta La Milia
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Maria Vittoria Grassini
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Section of Gastroenterology & Hepatology, Department of Health Promotion Sciences Maternal and Infant Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, 90127 Palermo, Italy
| | - Nicola Pugliese
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Massimo De Giorgio
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Stefano Fagiuoli
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Gastroenterology, Department of Medicine, University of Milan Bicocca, 20126 Milan, Italy
| |
Collapse
|
239
|
Ito T, Hashimoto H, Kaku-Ito Y, Tanaka Y, Nakahara T. Nail Apparatus Melanoma: Current Management and Future Perspectives. J Clin Med 2023; 12:jcm12062203. [PMID: 36983205 PMCID: PMC10057171 DOI: 10.3390/jcm12062203] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Nail apparatus melanoma (NAM) is a rare type of cutaneous melanoma that belongs to the acral melanoma subtype. NAM is managed principally in accordance with the general treatment for cutaneous melanoma, but there is scarce evidence in support of this in the literature. Acral melanoma is genetically different from non-acral cutaneous melanoma, while recently accumulated data suggest that NAM also has a different genetic background from acral melanoma. In this review, we focus on recent advances in the management of NAM. Localized NAM should be surgically removed; amputation of the digit and digit-preserving surgery have been reported. Sentinel lymph node biopsy can be considered for invasive NAM for the purpose of accurate staging. However, it is yet to be clarified whether patients with metastatic sentinel lymph nodes can be safely spared completion lymph node dissection. Similar to cutaneous melanoma, immune checkpoint inhibitors and BRAF/MEK inhibitors are used as the first-line treatment for metastatic NAM, but data on the efficacy of these therapies remain scarce. The therapeutic effects of immune checkpoint inhibitors could be lower for NAM than for cutaneous melanoma. This review highlights the urgent need to accumulate data to better define the optimal management of this rare melanoma.
Collapse
Affiliation(s)
- Takamichi Ito
- Correspondence: ; Tel.: +81-92-642-5585; Fax: +81-92-642-5600
| | | | | | | | | |
Collapse
|
240
|
van Akkooi ACJ, Schadendorf D, Eggermont AMM. Alternatives and reduced need for sentinel lymph node biopsy (SLNB) staging for melanoma. Eur J Cancer 2023; 182:163-169. [PMID: 36681612 DOI: 10.1016/j.ejca.2022.12.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
Sentinel lymph node biopsy (SLNB) has been introduced in the 1990s to identify patients who might benefit from completion lymph node dissection. Neither SLNB nor CLND improved survival, but SLNB staging did provide the best staging, above Breslow thickness and ulceration. The SLN status and SLN tumour burden were used in all trials until date looking at modern adjuvant systemic therapy with immune checkpoint inhibition (ICI) or targeted therapies (TT). Adjuvant ICI and TT are shifting towards stage II melanoma. The question is whether there is still role for SLNB in melanoma, in this day and age, and if so, how does the future look for SLNB staging? The SLN status and SLN tumour burden might be useful for a consultation to discuss the number needed to treat in a risk/benefit discussion. For stage IIB/C patients, it seems likely, however, that patients will forego a nuclear scan followed by the risk of surgical intervention and morbidity associated with SLNB if they opt to receive adjuvant therapy regardless of the SLNB result. For stage I/IIA, it is still required to detect high-risk patients who might benefit from adjuvant therapy. However, biomarkers are emerging, such as gene expression profilers (GEP), immunohistological signatures and liquid biopsies with ctDNA. There still is a role for SLNB staging in melanoma today, but we expect that the availability of therapeutic option independent of SLNB status as well as emergence of validated biomarkers to predict risk will reduce the need for SLNB staging in the upcoming decade to the point it will no longer be used.
Collapse
Affiliation(s)
- Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia; Royal Prince Alfred Hospital, Sydney, Australia.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer DKTK Consortium, Partner Site, Germany; Research Center One Health Ruhr, Research Alliance Ruhr, University Duisburg-Essen, Duisburg, Germany
| | - Alexander M M Eggermont
- Comprehensive Cancer Center München, Technical University München & Ludwig Maximiliaan University, München, Germany; University Medical Center Utrecht & Princess Maxima Center, Utrecht, the Netherlands
| |
Collapse
|
241
|
Xia TY, Cakmakoglu C, Kwiecien GJ, Gastman BR. Prophylactic Lymphaticovenous Anastomosis Performed with Lymphadenectomy is Oncologically Safe for Melanoma. Ann Surg Oncol 2023; 30:1823-1829. [PMID: 36471187 DOI: 10.1245/s10434-022-12791-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND A major concern of lymphaticovenous anastomosis (LVA), which has not been studied, is increased risk of metastasis. Melanoma patients with macrometastatic lymph node disease represent a high-risk group for recurrence and metastasis. On the basis of a literature review, this present study is the first to evaluate the impact of prophylactic LVA on cancer survival and recurrence. METHODS This was a comparison study of patients with cutaneous melanoma who underwent therapeutic lymphadenectomy alone (comparison group) or combined with prophylactic LVA (LVA group) between 2014 and 2020. A single surgeon performed all cancer resections, therapeutic lymphadenectomies, and LVA. Exclusion criteria included non-melanoma skin cancers, stage IV cancers before lymphadenectomy, microscopic lymphatic disease (i.e., positive sentinel node biopsy was the sole indication for lymph node dissection), or follow-up time less than 12 months unless the patient died earlier owing to melanoma-related complications. RESULTS This study included 23 patients in the LVA group and 22 consecutive patients in the comparison group. The two groups were similar in age, sex, and cancer stages. The comparison group had longer follow-up times (median 67.62 versus 29.73 months in the LVA group; p < 0.01). Average size of largest metastatic lymph node was 45.91 ± 35.03 mm and 44.54 ± 23.32 mm in the LVA and comparison groups, respectively (p = 0.99). There were no differences in OS, DMFS, and RFS times after more than 2 years of follow-up since the index surgery. CONCLUSION Prophylactic LVA performed for macrometastatic melanoma is not a strong risk factor for relapse and metastasis. LEVEL OF EVIDENCE II Therapeutic.
Collapse
Affiliation(s)
- Thomas Y Xia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Cagri Cakmakoglu
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Brian R Gastman
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
242
|
Jiminez V, Yusuf N. Role of the Microbiome in Immunotherapy of Melanoma. Cancer J 2023; 29:70-74. [PMID: 36957976 DOI: 10.1097/ppo.0000000000000648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Novel immunotherapeutics for advanced melanoma have drastically changed survival rates and management strategies in recent years. Immune checkpoint inhibitors have emerged as efficacious agents for some patients but have not been proven to be as beneficial in other patient cohorts. Recent investigation into this observation has implicated the gut microbiome as a potential immunomodulator in regulating patient response to therapy. Numerous studies have provided evidence for this link. Bacterial colonization patterns have been associated with therapeutic outcomes, under the notion that favorable commensal organisms improve host immune response. This review aims to report the most recent and pertinent findings related to the relationship between gut microbial communities and melanoma therapy efficacy. This article also highlights the emerging frontier of artificial intelligence in its application regarding patient microbial composition evaluation, predictive models for therapy response, and recommendations for the future of probiotics and dietary interventions to optimize melanoma survival and outcomes.
Collapse
Affiliation(s)
| | - Nabiha Yusuf
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
243
|
Patient Preferences and Satisfaction With Decisions in Stage-III Melanoma: A Mixed Methods Study. J Surg Res 2023; 283:485-493. [PMID: 36436284 DOI: 10.1016/j.jss.2022.10.079] [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: 06/30/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Rapid accumulation of data in surgical and medical oncology has changed the treatment landscape for patients with stage-III melanoma, introducing options for active surveillance and adjuvant systemic therapy; however, these options have increased the complexity of decision making. METHODS We conducted an explanatory sequential mixed-methods study consisting of surveys and semistructured interviews among patients diagnosed with stage-III melanoma at a single institution from August 2019 to December 2021. The survey included the validated 30-point satisfaction with decision scale (SWD). The interview guide was developed using a shared decision-making framework. RESULTS Twenty-six participants completed the survey (response rate 40%) and 17 were interviewed. In the survey, 69% of participants reported receiving a recommendation for active surveillance and 23% received a recommendation for adjuvant systemic therapy. Overall SWD for treatment of the lymph node basin and adjuvant systemic therapy was high at 27.94 and 26.21 out of 30, respectively. In the interviews, participants stressed the importance of the physician's recommendation as well as the desire to minimize intervention and avoid potential side effects in their decisions. However, they demonstrated persistent knowledge gaps in their understanding of the treatment options. CONCLUSIONS Like other cancer types where the option for active surveillance exists, the physician's recommendation is influential in shaping decisions for patients with stage-III melanoma. Physicians can improve shared decision making in this complex treatment landscape through improved multidisciplinary collaboration and mechanisms for ensuring patients' understanding of the treatment options.
Collapse
|
244
|
Verzijl CRC, van de Peppel IP, Eilers RE, Bloks VW, Wolters JC, Koehorst M, Kloosterhuis NJ, Havinga R, Jalving M, Struik D, Jonker JW. Pharmacological inhibition of MEK1/2 signaling disrupts bile acid metabolism through loss of Shp and enhanced Cyp7a1 expression. Biomed Pharmacother 2023; 159:114270. [PMID: 36680812 DOI: 10.1016/j.biopha.2023.114270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
The RAS-MAPK signaling pathway is one of the most frequently dysregulated pathways in human cancer. Small molecule inhibitors directed against this pathway have clinical activity in patients with various cancer types and can improve patient outcomes. However, the use of these drugs is associated with adverse effects, which can result in dose reduction or treatment interruption. A better molecular understanding of on-target, off-tumor effects may improve toxicity management. In the present study, we aimed to identify early initiating biological changes in the liver upon pharmacological inhibition of the RAS-MAPK signaling pathway. To this end, we tested the effect of MEK inhibitor PD0325901 using mice and human hepatocyte cell lines. Male C57BL/6 mice were treated with either vehicle or PD0325901 for six days, followed by transcriptome analysis of the liver and phenotypic characterization. Pharmacological MEK inhibition altered the expression of 423 genes, of which 78 were upregulated and 345 were downregulated. We identified Shp, a transcriptional repressor, and Cyp7a1, the rate-limiting enzyme in converting cholesterol to bile acids, as the top differentially expressed genes. PD0325901 treatment also affected other genes involved in bile acid regulation, which was associated with changes in the composition of plasma bile acids and composition and total levels of fecal bile acids and elevated predictive biomarkers of early liver toxicity. In conclusion, short-term pharmacological MEK inhibition results in profound changes in bile acid metabolism, which may explain some of the clinical adverse effects of pharmacological inhibition of the RAS-MAPK pathway, including gastrointestinal complications and hepatotoxicity.
Collapse
Affiliation(s)
- Cristy R C Verzijl
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ivo P van de Peppel
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roos E Eilers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 Groningen, GZ, The Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dicky Struik
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
245
|
Berania I, Tzelnick S, de Almeida JR, McKinnon G, Goldstein DP. Practice patterns for positive sentinel lymph node in head and neck melanoma. Head Neck 2023; 45:555-560. [PMID: 36513594 DOI: 10.1002/hed.27262] [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: 04/02/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND An international survey was conducted to investigate the preferences for completion lymph node dissection (CLND) in head and neck melanomas. METHODS A questionnaire was sent through the American Society of Head & Neck Surgery (AHNS) and Canadian Society of Otolaryngology-Head and Neck Surgery (CSO). RESULTS Hundred and forty-nine surgeons completed the survey. Response rate was 6.3% and 9.7% from AHNS and CSO members, respectively. When presented the scenario of a 47-year-old male with a clinical T3bN0 cheek melanoma, with 1/1 positive sentinel lymph node (SLN) with nodal deposit <2 mm, 72 of respondents (48.3%) would perform a CLND. Reasons for CLND included multiples positive SLN (64.1%), size of nodal deposits (54.2%), and perceived lack of compliance to follow-up (54.2%). Surgeons with access to immunotherapy treatment were less likely to recommend CLND (p = 0.025). CONCLUSIONS Following SLN biopsy, nearly half of the surveyed head and neck surgeons would recommend CLND, which contrasts with the current melanoma practice patterns in other anatomic locations. However, compared with an earlier study in the literature it does seem that there has been a shift away from completion neck dissection. Further investigation into understanding practice variations is warranted.
Collapse
Affiliation(s)
- Ilyes Berania
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sharon Tzelnick
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - John R de Almeida
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gregory McKinnon
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head and Neck Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
246
|
Egeler MD, van Leeuwen M, Fraterman I, van den Heuvel NMJ, Boekhout AH, Lai-Kwon J, Wilthagen EA, Eriksson H, Haanen JB, Wilgenhof S, Ascierto PA, van Akkooi ACJ, van de Poll-Franse LV. Common toxicities associated with immune checkpoint inhibitors and targeted therapy in the treatment of melanoma: A systematic scoping review. Crit Rev Oncol Hematol 2023; 183:103919. [PMID: 36736511 DOI: 10.1016/j.critrevonc.2023.103919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/23/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION This systematic scoping review compares the toxicities experienced by patients receiving immune checkpoint inhibitors (ICIs) or targeted therapy (TT) for stage III (resected and unresectable) and stage IV melanoma. METHODS OVID Medline, Embase, and PsycInfo were searched to identify Phase III trials reporting toxicities of FDA-approved ICIs and TT for advanced melanoma. AEs that were reported by ≥ 10% of patients in the evaluated trials were included. RESULTS Toxicity profiles of 11208 patients from 24 studies were reviewed. The rate of AEs was lower with ICIs compared to TT. However, ICIs were associated with higher rates of long-term or permanent AEs compared to TT, where toxicities generally were shortterm and reversible with treatment discontinuation. CONCLUSION The toxicity profiles of ICIs and TT vary substantially. Whilst the rate of AEs was lower with ICIs than during TT, it was also associated with higher rates of potentially chronic AEs.
Collapse
Affiliation(s)
- Mees D Egeler
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Marieke van Leeuwen
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Itske Fraterman
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Noelle M J van den Heuvel
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annelies H Boekhout
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Julia Lai-Kwon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Erica A Wilthagen
- Scientific Information Service, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical Unit Head-Neck-, Lung-, Skin Cancer, Skin Cancer Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - John B Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Lonneke V van de Poll-Franse
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands; Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic diseases (CoRPS), Tilburg University, Tilburg, the Netherlands
| |
Collapse
|
247
|
Abstract
LEARNING OBJECTIVES After reading this article and viewing the videos, the participant should be able to: 1. Discuss margins for in situ and invasive disease and describe reconstructive options for wide excision defects, including the keystone flap. 2. Describe a digit-sparing alternative for subungual melanoma. 3. Calculate personalized risk estimates for sentinel node biopsy using predictive nomograms. 4. Describe the indications for lymphadenectomy and describe a technique intended to reduce the risk of lymphedema following lymphadenectomy. 5. Offer options for in-transit melanoma management. SUMMARY Melanoma management continues to evolve, and plastic surgeons need to stay at the forefront of advances and controversies. Appropriate margins for in situ and invasive disease require consideration of the trials on which they are based. A workhorse reconstruction option for wide excision defects, particularly in extremities, is the keystone flap. There are alternative surgical approaches to subungual tumors besides amputation. It is now possible to personalize a risk estimate for sentinel node positivity beyond what is available for groups of patients with a given stage of disease. Sentinel node biopsy can be made more accurate and less morbid with novel adjuncts. Positive sentinel node biopsies are now rarely managed with completion lymphadenectomy. Should a patient require lymphadenectomy, immediate lymphatic reconstruction may mitigate the lymphedema risk. Finally, there are minimally invasive modalities for effective control of in-transit recurrences.
Collapse
|
248
|
Tan EY, Pazdirkova M, Taylor AJ, Singh N, Iyer GR. Evaluation of a Low-Fat Low-Calorie Meal on the Relative Bioavailability of Trametinib and Dabrafenib: Results From a Randomized, Open-Label, 2-Part Study in Healthy Participants. Clin Pharmacol Drug Dev 2023; 12:333-342. [PMID: 36662829 DOI: 10.1002/cpdd.1220] [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: 08/08/2022] [Accepted: 12/19/2022] [Indexed: 01/21/2023]
Abstract
In this randomized, open-label, 2-part, 2 × 2 crossover, phase 1 study, the effect of a low-fat low-calorie (LFLC) meal on the relative bioavailability of a trametinib 2-mg tablet or dabrafenib 150-mg capsule was evaluated in healthy participants. Trametinib adjusted geometric mean ratios (90%CI) of fed : fasted for area under the concentration-time curve (AUC) from time 0 to the last quantifiable concentration and AUC from time 0 extrapolated to infinity were 0.76 (0.71-0.82) and 0.82 (0.77-0.88), respectively. For dabrafenib, the adjusted geometric mean ratios of AUC from time 0 to the last quantifiable concentration and AUC from time 0 extrapolated to infinity (90%CI) for fed:fasted were 0.85 (0.79-0.91) and 0.86 (0.80-0.92), respectively. Consumption of an LFLC meal delayed trametinib and dabrafenib absorption, with an increase in time to maximum concentration of ≈15 and ≈30 minutes, respectively, compared to the fasted state. These findings indicate that consumption of an LFLC meal reduced the bioavailability and delayed the absorption of trametinib and dabrafenib, supporting current recommendations to administer both drugs in the fasting state; however, an occasional LFLC meal is unlikely to affect the pharmacokinetics of the drugs once steady state is reached and, by consequence, not likely to alter the overall intended efficacy.
Collapse
Affiliation(s)
- Eugene Y Tan
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Amanda J Taylor
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Namrata Singh
- Novartis Healthcare Private Limited, Hyderabad, India
| | - Ganesh R Iyer
- Novartis Institute of Biomedical Research Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
249
|
de Meza MM, Blokx WAM, Bonenkamp HJ, Blank CU, Aarts MJB, van den Berkmortel FWPJ, Boers-Sonderen MJ, de Groot JWB, Haanen JB, Hospers GAP, Kapiteijn EW, van Not OJ, Piersma D, van Rijn RS, Stevense-Den Boer MA, van der Veldt AAM, Vreugdenhil G, van den Eertwegh AJM, Suijkerbuijk KPM, Wouters MWJM. Adjuvant treatment of in-transit melanoma: Narrowing the knowledge gap left by clinical trials. Int J Cancer 2023; 153:389-398. [PMID: 36843260 DOI: 10.1002/ijc.34485] [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: 05/02/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 02/28/2023]
Abstract
Few clinical trials address efficacy of adjuvant systemic treatment in patients with in-transit melanoma (ITM). This study describes adjuvant systemic therapy of ITM patients beyond clinical trials. In this study, we included stage III adjuvant-treated melanoma patients registered in the nationwide Dutch Melanoma Treatment Registry between July 2018 and December 2020. Patients were divided into three groups: nodal disease only, ITM only and ITM and nodal disease. Recurrence patterns, recurrence-free survival (RFS) and overall survival (OS) at 12-months were analyzed. In our study population of 1037 patients, 66.8% had nodal disease only, 16.7% had ITM only and 16.2% had ITM with nodal disease. RFS at 12-months was comparable in the nodal only and ITM only group (72.2% vs70.1%, P = .97) but lower in ITM and nodal disease patients (57.8%; P = .01, P < .01). Locoregional metastases occurred as first recurrence in 38.9% nodal disease only, 71.9% of ITM-only and 44.0% of ITM and nodal disease patients. Distant recurrences occurred in 42.3%, 18.8% and 36.0%, respectively (P = .02). 12-months OS was not significantly different for nodal disease only patients compared with ITM-only (94.4% vs 97.6%, P = .06) but was significantly higher for ITM-only compared with ITM and nodal disease patients (97.6% vs 91.0%, P < .01). In conclusion, we showed that in the adjuvant setting, RFS rates in ITM-only patients are similar to non-ITM, though better than in ITM and nodal disease patients. Adjuvant-treated ITM-only patients less often experience distant recurrences and have a superior OS compared with ITM and nodal disease patients.
Collapse
Affiliation(s)
- Melissa M de Meza
- Dutch Institute for Clinical Auditing, Leiden, The Netherlands.,Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Han J Bonenkamp
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cristian U Blank
- Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - John B Haanen
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Olivier J van Not
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Rozemarijn S van Rijn
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | | | - Astrid A M van der Veldt
- Departments of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gerard Vreugdenhil
- Department of Internal Medicine, Maxima Medical Center, Eindhoven, The Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Michel W J M Wouters
- Dutch Institute for Clinical Auditing, Leiden, The Netherlands.,Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands.,Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
250
|
Gouda M, Subbiah V. Precision oncology for BRAF-mutant cancers with BRAF and MEK inhibitors: from melanoma to tissue-agnostic therapy. ESMO Open 2023; 8:100788. [PMID: 36842301 PMCID: PMC9984800 DOI: 10.1016/j.esmoop.2023.100788] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 02/26/2023] Open
Abstract
BRAF activation occurs as part of the mitogen-activated protein kinase (MAPK) cellular signaling pathway which leads to increased cellular proliferation and survival. Mutations in BRAF can result in unbridled activation of downstream kinases with subsequent uncontrolled cellular growth that formulate the basis for oncogenesis in multiple tumor types. Targeting BRAF by selective inhibitors has been one of the early successes in precision oncology. Agents have been explored either as monotherapy or in combination with MEK inhibition in BRAF V600-mutant pan-cancers and with EGFR inhibition in colorectal cancer. Spectrum of BRAF inhibition has evolved from being melanoma-specific to being a pan-cancer target. In this article, we review BRAF and MEK inhibitor drug development journey from tissue-specific melanoma, non-small-cell lung cancer, and anaplastic thyroid cancer to tissue-agnostic approvals.
Collapse
Affiliation(s)
- M.A. Gouda
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA,Department of Clinical Oncology, Faculty of Medicine, Menoufia University, Shebin Al-Kom, Menoufia, Egypt
| | - V. Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA,Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston,MD Anderson Cancer Network, The University of Texas MD Anderson Cancer Center, Houston, USA,Correspondence to: Prof. Vivek Subbiah, Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA. Tel: +1-713-563-1930 @VivekSubbiah
| |
Collapse
|