351
|
Villani A, Potestio L, Fabbrocini G, Troncone G, Malapelle U, Scalvenzi M. The Treatment of Advanced Melanoma: Therapeutic Update. Int J Mol Sci 2022; 23:ijms23126388. [PMID: 35742834 PMCID: PMC9223461 DOI: 10.3390/ijms23126388] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Cutaneous melanoma is the main cause of death for skin cancer. The majority of patients with a diagnosis of melanoma have localized disease, which can be successfully treated with surgical treatment. However, the surgical approach is not curative for advanced melanoma (AM). Indeed, the management of AM is still challenging, since melanoma is the solid tumor with the highest number of mutations and cancer cells have the capacity to evade the immune system. In the past, the treatment of AM relied on chemotherapeutic agents, without showing efficacy data. Recent knowledge on melanoma pathogenesis as well as the introduction of immunotherapies, targeted therapies vaccines, small molecules, and combination therapies has revolutionized AM management, showing promising results in terms of effectiveness and safety. The aim of this review is to assess and to discuss the role of emerging therapies for AM management in order to obtain a complete overview of the currently available treatment options and future perspectives.
Collapse
Affiliation(s)
- Alessia Villani
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (L.P.); (G.F.); (M.S.)
- Correspondence: ; Tel.: +39-081-7462457; Fax: +39-081-7462442
| | - Luca Potestio
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (L.P.); (G.F.); (M.S.)
| | - Gabriella Fabbrocini
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (L.P.); (G.F.); (M.S.)
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (G.T.); (U.M.)
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (G.T.); (U.M.)
| | - Massimiliano Scalvenzi
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (L.P.); (G.F.); (M.S.)
| |
Collapse
|
352
|
Dimitriou F, Hauschild A, Mehnert JM, Long GV. Double Trouble: Immunotherapy Doublets in Melanoma-Approved and Novel Combinations to Optimize Treatment in Advanced Melanoma. Am Soc Clin Oncol Educ Book 2022; 42:1-22. [PMID: 35658500 DOI: 10.1200/edbk_351123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Immune checkpoint inhibitors, particularly anti-PD-1-based immune checkpoint inhibitors, have dramatically improved outcomes for patients with advanced melanoma and are currently deemed a standard of care. Ipilimumab/nivolumab is the first combination of immune checkpoint inhibitors to improve progression-free survival and overall survival in the first-line setting, with durable responses and the longest median overall survival, 72.1 months, of any drug therapy approved for advanced melanoma. However, its use is limited by the high rate of severe (grade 3-4) treatment-related adverse events. More recently, the novel immune checkpoint inhibitor combination of nivolumab/relatlimab (anti-PD-1/anti-LAG3) showed improved progression-free survival compared with nivolumab alone in the first-line setting and was well tolerated; thus, it is likely this combination will be added to the armamentarium as a first-line treatment for advanced melanoma. These changes in the treatment landscape have several treatment implications for decision-making. The choice of first-line systemic drug therapy, and the decision between immune checkpoint inhibitor monotherapy or combination therapy, requires a comprehensive assessment of disease-related factors and patient characteristics. Despite this striking progress, many patients' disease still progresses. Several new agents and therapeutic approaches are under investigation in clinical trials. Intralesional treatments hold promise for accessible metastases, although their broad application in the clinic will be limited. Prognostic and predictive biomarkers, as well as strategies to reduce treatment-related toxicities and overcome resistance, are required and are now the focus of clinical and translational research.
Collapse
Affiliation(s)
- Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Janice M Mehnert
- NYU Grossman School of Medicine and Perlmutter Cancer Center, New York, NY
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| |
Collapse
|
353
|
Lao CD, Khushalani NI, Angeles C, Petrella TM. Current State of Adjuvant Therapy for Melanoma: Less Is More, or More Is Better? Am Soc Clin Oncol Educ Book 2022; 42:1-7. [PMID: 35658502 DOI: 10.1200/edbk_351153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advances in melanoma treatments over the past decade have changed the course of survival for patients. Several adjuvant therapies have been approved and are now considered standard of care for high-risk patients. These therapies have shown improvements for recurrence-free survival and distant metastases-free survival, but not overall survival, as the data are maturing. The 5-year recurrence-free survival in the COMBI-AD study, which compared dabrafenib and trametinib with placebo, was 65% and 58%, respectively. In the KEYNOTE-054 study, the recurrence-free survival at 3 years was 63.7% versus 41%. Despite these advances, approximately 50% of patients will succumb to their disease. Adjuvant therapy is considered potentially curative and avoids the morbidity of relapsed disease and the poor outcomes seen in metastatic disease. However, the lack of overall survival benefit in clinical trials of patients with high-risk stage II and stage III disease raises the question of whether it is more efficacious to treat when there is residual microscopic disease, or to wait until the disease recurs to avoid treating those who may have been cured by surgery alone. Immunotherapy also has the potential for substantial toxicity that may be lifelong; hence, discussion of risks and benefits of therapy is warranted because there should be less tolerance for substantial toxicity in the adjuvant setting. Adjuvant trials are needed that will integrate biomarkers to allow for better selection of patients who will truly benefit from adjuvant therapy.
Collapse
Affiliation(s)
| | | | | | - Teresa M Petrella
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
354
|
The role of sentinel node tumor burden in modeling the prognosis of melanoma patients with positive sentinel node biopsy: an Italian melanoma intergroup study (N = 2,086). BMC Cancer 2022; 22:610. [PMID: 35659273 PMCID: PMC9166524 DOI: 10.1186/s12885-022-09705-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/26/2022] [Indexed: 12/25/2022] Open
Abstract
Background The management of melanoma patients with metastatic melanoma in the sentinel nodes (SN) is evolving based on the results of trials questioning the impact of completion lymph node dissection (CLND) and demonstrating the efficacy of new adjuvant treatments. In this landscape, new prognostic tools for fine risk stratification are eagerly sought to optimize the therapeutic path of these patients. Methods A retrospective cohort of 2,086 patients treated with CLND after a positive SN biopsy in thirteen Italian Melanoma Centers was reviewed. Overall survival (OS) was the outcome of interest; included independent variables were the following: age, gender, primary melanoma site, Breslow thickness, ulceration, sentinel node tumor burden (SNTB), number of positive SN, non-sentinel lymph nodes (NSN) status. Univariate and multivariate survival analyses were performed using the Cox proportional hazard regression model. Results The 3-year, 5-year and 10-year OS rates were 79%, 70% and 54%, respectively. At univariate analysis, all variables, except for primary melanoma body site, were found to be statistically significant prognostic factors. Multivariate Cox regression analysis indicated that older age (P < 0.0001), male gender (P = 0.04), increasing Breslow thickness (P < 0.0001), presence of ulceration (P = 0.004), SNTB size (P < 0.0001) and metastatic NSN (P < 0.0001) were independent negative predictors of OS. Conclusion The above results were utilized to build a nomogram in order to ease the practical implementation of our prognostic model, which might improve treatment personalization.
Collapse
|
355
|
Landström FJ, Jakobsson FE, Kristiansson SJ. Successful electrochemotherapy treatment of a large bleeding lymph node melanoma metastasis. Melanoma Manag 2022; 9:MMT61. [PMID: 35813184 PMCID: PMC9260494 DOI: 10.2217/mmt-2021-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 06/09/2022] [Indexed: 12/21/2022] Open
Abstract
Despite the progress in immunotherapy and targeted therapy for patients with cutaneous malignant melanoma not all patients with loco-regional recurrences will respond to treatment. Electrochemotherapy is a relatively new treatment modality where the efficacy of a chemotherapeutic drug is enhanced by an electrical field. Here we report a case of a 68-year-old woman with a large therapy resistant inguinal lymph node melanoma metastasis complicated by bleeding that was successfully treated with electrochemotherapy.
Collapse
Affiliation(s)
- Fredrik J Landström
- Department of Otolaryngology, Örebro University Hospital, Örebro, Sweden.,Örebro University, Örebro, Sweden
| | | | | |
Collapse
|
356
|
Mousavi I, Olivier T, Prasad V. Cost per Event Averted in Cancer Trials in the Adjuvant Setting From 2018 to 2022. JAMA Netw Open 2022; 5:e2216058. [PMID: 35687338 PMCID: PMC9187950 DOI: 10.1001/jamanetworkopen.2022.16058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/04/2022] [Indexed: 01/12/2023] Open
Abstract
Importance Adjuvant therapies are often approved based on improvements in disease-, progression-, or relapse-free survival (ie, an event). An important estimate in adjuvant therapies is the cost per event averted. Objective To characterize the costs per event averted of anticancer drugs approved by the US Food and Drug Administration (FDA) between January 2018 and March 2022. Design, Setting, and Participants In this cross-sectional study, all approvals were reviewed from the FDA website. Approvals selected for the analysis needed to be drawn from randomized trials involving anticancer drugs studied in the adjuvant treatment of solid tumors. Treatments of nonsolid tumors; nonrandomized, noncontrolled trials; and unpublished trials were excluded. Approvals between January 2018 and March 2022 were included, and 11 trials met inclusion criteria. The monthly costs of each agent were abstracted from the Micromedex RED BOOK database. All variables included in calculations were derived from the original trial publication. Information abstracted for each approval included the name of the drug approved, name of the trial, primary end point, dosing regimen in the trial, median duration of treatment, tumor type, monthly costs per drug, and the number needed to treat for the primary end point. Data were analyzed in March 2022. Main Outcomes and Measures The cost per event averted for each agent studied in each clinical trial; the monthly cost of each drug, the cost per patient for each drug, the primary clinical end point studied in each trial; the study's design and setting. Results A total of 11 approvals were included in the study. From January 2018 to March 2022, all approvals were based on a surrogate end point as the primary end point, with no trials demonstrating, to date, an overall survival benefit. The median cost per event averted of drugs in the adjuvant setting was $1 610 000 (range, $820 000 to $2 640 000). The median cost of a complete adjuvant treatment was $158 000 per patient. Conclusions and Relevance In this cross-sectional study, anticancer agents in the adjuvant setting were approved based on disease-, progression-, event-, or relapse-free survival. Costs per event averted were high. These findings suggest that these drugs may not achieve wide or global use unless their efficacy improves or their prices decrease.
Collapse
Affiliation(s)
- Idine Mousavi
- School of Medicine, New York Medical College, Valhalla
| | - Timothée Olivier
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco
| |
Collapse
|
357
|
Mulder EEAP, Verver D, van der Klok T, de Wijs CJ, van den Bosch TPP, De Herdt MJ, van der Steen B, Verhoef C, van der Veldt AAM, Grünhagen DJ, Koljenovic S. Mesenchymal-epithelial transition factor (MET) immunoreactivity in positive sentinel nodes from patients with melanoma. Ann Diagn Pathol 2022; 58:151909. [PMID: 35151198 DOI: 10.1016/j.anndiagpath.2022.151909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVE Patients with cutaneous melanoma and a positive sentinel node (SN) are currently eligible for adjuvant treatment with targeted therapy and immune checkpoint inhibitors. Near-infrared (NIR) fluorescence imaging could be an alternative and less invasive tool for SN biopsy to select patients for adjuvant treatment. One potential target for NIR is the mesenchymal-epithelial transition factor (MET). This study aimed to assess MET immunoreactivity in positive SNs and to evaluate its potential diagnostic, prognostic and therapeutic value. METHODS In this retrospective study, positive SN samples from patients with primary cutaneous melanoma were collected to assess MET immunoreactivity. To this end, paraffin-embedded SNs were stained for MET (monoclonal antibody D1C2). A 4-point Histoscore was used to determine cytoplasmic and membranous immunoreactivity (0 negative/1 weak/2 moderate/3 strong). Samples were considered positive when ≥10% of the cancer cells showed MET expression (staining intensity ≥1). Patient and clinicopathological characteristics were used for descriptive statistics, binary logistic regression, and survival analyses. RESULTS Positive MET immunohistochemistry was observed in 24 out of 37 samples (65%). No statistically significant associations were found between MET positivity and the following prognostic factors: Breslow thickness (P = 0.961), ulceration (P = 1.000), and SN tumor burden (P = 0.792). According to MET positivity, Kaplan-Meier curves showed no significant differences in survival. CONCLUSION This exploratory study found no evidence to support MET immunoreactivity in positive SNs as a possible diagnostic or prognostic indicator in patients with melanoma.
Collapse
Affiliation(s)
- Evalyn E A P Mulder
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Daniëlle Verver
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | | | - Calvin J de Wijs
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | | | - Maria J De Herdt
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Berdine van der Steen
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | | |
Collapse
|
358
|
van Akkooi ACJ, Hieken TJ, Burton EM, Ariyan C, Ascierto PA, Asero SVMA, Blank CU, Block MS, Boland GM, Caraco C, Chng S, Davidson BS, Duprat Neto JP, Faries MB, Gershenwald JE, Grunhagen DJ, Gyorki DE, Han D, Hayes AJ, van Houdt WJ, Karakousis GC, Klop WMC, Long GV, Lowe MC, Menzies AM, Olofsson Bagge R, Pennington TE, Rutkowski P, Saw RPM, Scolyer RA, Shannon KF, Sondak VK, Tawbi H, Testori AAE, Tetzlaff MT, Thompson JF, Zager JS, Zuur CL, Wargo JA, Spillane AJ, Ross MI. Neoadjuvant Systemic Therapy (NAST) in Patients with Melanoma: Surgical Considerations by the International Neoadjuvant Melanoma Consortium (INMC). Ann Surg Oncol 2022; 29:3694-3708. [PMID: 35089452 DOI: 10.1245/s10434-021-11236-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022]
Abstract
Exciting advances in melanoma systemic therapies have presented the opportunity for surgical oncologists and their multidisciplinary colleagues to test the neoadjuvant systemic treatment approach in high-risk, resectable metastatic melanomas. Here we describe the state of the science of neoadjuvant systemic therapy (NAST) for melanoma, focusing on the surgical aspects and the key role of the surgical oncologist in this treatment paradigm. This paper summarizes the past decade of developments in melanoma treatment and the current evidence for NAST in stage III melanoma specifically. Issues of surgical relevance are discussed, including the risk of progression on NAST prior to surgery. Technical aspects, such as the definition of resectability for melanoma and the extent and scope of routine surgery are presented. Other important issues, such as the utility of radiographic response evaluation and method of pathologic response evaluation, are addressed. Surgical complications and perioperative management of NAST related adverse events are considered. The International Neoadjuvant Melanoma Consortium has the goal of harmonizing NAST trials in melanoma to facilitate rapid advances with new approaches, and facilitating the comparison of results across trials evaluating different treatment regimens. Our ultimate goals are to provide definitive proof of the safety and efficacy of NAST in melanoma, sufficient for NAST to become an acceptable standard of care, and to leverage this platform to allow more personalized, biomarker-driven, tailored approaches to subsequent treatment and surveillance.
Collapse
Affiliation(s)
| | | | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Christian U Blank
- Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | | | - Corrado Caraco
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Sydney Chng
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Mark B Faries
- The Angeles Clinic, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - David E Gyorki
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dale Han
- Oregon Health and Science University, Portland, Oregon, USA
| | | | - Winan J van Houdt
- Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Willem M C Klop
- Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - 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, St. Leonards, NSW, Australia
- The Mater Hospital, North Sydney, NSW, Australia
| | - Michael C Lowe
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - 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, St. Leonards, NSW, Australia
- The Mater Hospital, North Sydney, NSW, Australia
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas E Pennington
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | | | - Hussein Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mike T Tetzlaff
- University of California San Francisco (UCSF), San Francisco, CA, USA
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Mater Hospital, North Sydney, NSW, Australia
| | | | - Charlotte L Zuur
- Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jennifer A Wargo
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Spillane
- 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, St. Leonards, NSW, Australia
- The Mater Hospital, North Sydney, NSW, Australia
| | - Merrick I Ross
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
359
|
Attrill GH, Owen CN, Ahmed T, Vergara IA, Colebatch AJ, Conway JW, Nahar KJ, Thompson JF, Pires da Silva I, Carlino MS, Menzies AM, Lo S, Palendira U, Scolyer RA, Long GV, Wilmott JS. Higher proportions of CD39+ tumor-resident cytotoxic T cells predict recurrence-free survival in patients with stage III melanoma treated with adjuvant immunotherapy. J Immunother Cancer 2022; 10:e004771. [PMID: 35688560 PMCID: PMC9189855 DOI: 10.1136/jitc-2022-004771] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Adjuvant immune checkpoint inhibitor (ICI) immunotherapies have significantly reduced the recurrence rate in high-risk patients with stage III melanoma compared with surgery alone. However, 48% of anti-PD-1-treated patients will develop recurrent disease within 4 years. There is a need to identify biomarkers of recurrence after adjuvant ICI to enable identification of patients in need of alternative treatment strategies. As cytotoxic T cells are critical for the antitumor response to anti-PD-1, we sought to determine whether specific subsets were predictive of recurrence in anti-PD-1-treated high-risk patients with stage III melanoma. METHODS Associations with recurrence in patients with stage III melanoma were sought by analyzing resection specimens (n=103) taken prior to adjuvant nivolumab/pembrolizumab±low-dose/low-interval ipilimumab. Multiplex immunohistochemistry was used to quantify intratumoral CD8+ T-cell populations using phenotypical markers CD39, CD103, and PD-1. RESULTS With a median follow-up of 19.3 months, 37/103 (36%) of patients had a recurrence. Two CD8+ T-cell subpopulations were significantly associated with recurrence. First, CD39+ tumor-resident memory cells (CD39+CD103+PD-1+CD8+ (CD39+ Trm)) comprised a significantly higher proportion of CD8+ T cells in recurrence-free patients (p=0.0004). Conversely, bystander T cells (CD39-CD103-PD-1-CD8+) comprised a significantly greater proportion of T cells in patients who developed recurrence (p=0.0002). Spatial analysis identified that CD39+ Trms localized significantly closer to melanoma cells than bystander T cells. Multivariable analysis confirmed significantly improved recurrence-free survival (RFS) in patients with a high proportion of intratumoral CD39+ Trms (1-year RFS high 78.1% vs low 49.9%, HR 0.32, 95% CI 0.15 to 0.69), no complete lymph node dissection performed, and less advanced disease stage (HR 2.85, 95% CI 1.13 to 7.19, and HR 1.29, 95% CI 0.59 to 2.82). The final Cox regression model identified patients who developed recurrence with an area under the curve of 75.9% in the discovery cohort and 69.5% in a separate validation cohort (n=33) to predict recurrence status at 1 year. CONCLUSIONS Adjuvant immunotherapy-treated patients with a high proportion of CD39+ Trms in their baseline melanoma resection have a significantly reduced risk of melanoma recurrence. This population of T cells may not only represent a biomarker of RFS following anti-PD-1 therapy, but may also be an avenue for therapeutic manipulation and enhancing outcomes for immunotherapy-treated patients with cancer.
Collapse
Affiliation(s)
- Grace Heloise Attrill
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Carina N Owen
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- The University of Bristol, Bristol Cancer Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Tasnia Ahmed
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J Colebatch
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | - Jordan W Conway
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kazi J Nahar
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital; Mater Hospital, Sydney, New South Wales, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Serigne Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Umaimainthan Palendira
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
360
|
Coulie J, Boon L, Vikkula M. Molecular Pathways and Possible Therapies for Head and Neck Vascular Anomalies. J Oral Pathol Med 2022; 51:878-887. [PMID: 35610188 DOI: 10.1111/jop.13318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Vascular Anomalies are a heterogenous group of vascular lesions that can be divided, according to the International Society for the Study of Vascular Anomalies Classification, into two main groups : Vascular Tumors and Vascular Malformations. Vascular Malformations can be further subdivided into slow-flow and fast-flow malformations. This clinical and radiological classification allows for a better understanding of vascular anomalies and aims to offer a more precise final diagnosis. Correct diagnosis is essential to propose the best treatment, which traditionally consists of surgery, embolization or sclerotherapy. Since a few years, medical treatment has become an important part of multidisciplinary treatment. Genetic and molecular knowledge of vascular anomalies are increasing rapidly and opens the door for a molecular classification of vascular anomalies according to the underlying pathways involved. The main pathways seem to be: PI3K/AKT/mTOR (PIKopathies) and RAS/RAF/MEK/ERK (RASopathies). Knowing the underlying molecular cascades allows us to use targeted medical therapies. The first part of this article aims to review the vascular anomalies seen in the head and neck region and their underlying molecular causes and involved pathways. The second part will propose an overview of the available targeted therapies based on the affected molecular cascade. This article summarizes theragnostic treatments available in vascular anomalies.
Collapse
Affiliation(s)
- Julien Coulie
- Center for Vascular Anomalies, Division of Plastic Surgery, VASCERN VASCA European Reference Centre, Saint Luc University Hospital, Brussels, Belgium.,Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Laurence Boon
- Center for Vascular Anomalies, Division of Plastic Surgery, VASCERN VASCA European Reference Centre, Saint Luc University Hospital, Brussels, Belgium.,Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Miikka Vikkula
- Center for Vascular Anomalies, Division of Plastic Surgery, VASCERN VASCA European Reference Centre, Saint Luc University Hospital, Brussels, Belgium.,Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| |
Collapse
|
361
|
Ascierto PA, Warner AB, Blank C, Caracò C, Demaria S, Gershenwald JE, Khushalani NI, Long GV, Luke JJ, Mehnert JM, Robert C, Rutkowski P, Tawbi HA, Osman I, Puzanov I. The "Great Debate" at Melanoma Bridge 2021, December 2nd-4th, 2021. J Transl Med 2022; 20:200. [PMID: 35538491 PMCID: PMC9087170 DOI: 10.1186/s12967-022-03406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/24/2022] [Indexed: 11/10/2022] Open
Abstract
The Great Debate session at the 2021 Melanoma Bridge virtual congress (December 2-4) featured counterpoint views from experts on seven important issues in melanoma. The debates considered the use of adoptive cell therapy versus use of bispecific antibodies, mitogen-activated protein kinase (MAPK) inhibitors versus immunotherapy in the adjuvant setting, whether the use of corticosteroids for the management of side effects have an impact on outcomes, the choice of programmed death (PD)-1 combination therapy with cytotoxic T-lymphocyte-associated antigen (CTLA)-4 or lymphocyte-activation gene (LAG)-3, whether radiation is needed for brain metastases, when lymphadenectomy should be integrated into the treatment plan and then the last debate, telemedicine versus face-to-face. As with previous Bridge congresses, the debates were assigned by meeting Chairs and positions taken by experts during the debates may not have necessarily reflected their respective personal view. Audiences voted 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.
| | | | | | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori "Fondazione Pascale" IRCCS, Naples, Italy
| | - Sandra Demaria
- Department of Radiation Oncology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia.,Faculty of Medicine & Health, The University of Sydney, Sydney, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Royal North Shore Hospital, Sydney, Australia
| | - Jason J Luke
- University of Pittsburgh Medical Center, UPMC) Hillman Cancer Center, Pittsburgh, PA, USA
| | - Janice M Mehnert
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Caroline Robert
- Institut de Cancérologie Gustave Roussy Et Université Paris-Saclay, Villejuif, France
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Hussein A Tawbi
- Melanoma Medical Oncology, Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Brain Metastasis Clinic, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Iman Osman
- New York University Langone Medical Center, New York, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
362
|
Thornton J, Chhabra G, Singh CK, Guzmán-Pérez G, Shirley CA, Ahmad N. Mechanisms of Immunotherapy Resistance in Cutaneous Melanoma: Recognizing a Shapeshifter. Front Oncol 2022; 12:880876. [PMID: 35515106 PMCID: PMC9066268 DOI: 10.3389/fonc.2022.880876] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/18/2022] [Indexed: 12/21/2022] Open
Abstract
Melanoma is one of the seven most common cancers in the United States, and its incidence is still increasing. Since 2011, developments in targeted therapies and immunotherapies have been essential for significantly improving overall survival rates. Prior to the advent of targeted and immunotherapies, metastatic melanoma was considered a death sentence, with less than 5% of patients surviving more than 5 years. With the implementation of immunotherapies, approximately half of patients with metastatic melanoma now survive more than 5 years. Unfortunately, this also means that half of the patients with melanoma do not respond to current therapies and live less than 5 years after diagnosis. One major factor that contributes to lower response in this population is acquired or primary resistance to immunotherapies via tumor immune evasion. To improve the overall survival of melanoma patients new treatment strategies must be designed to minimize the risk of acquired resistance and overcome existing primary resistance. In recent years, many advances have been made in identifying and understanding the pathways that contribute to tumor immune evasion throughout the course of immunotherapy treatment. In addition, results from clinical trials focusing on treating patients with immunotherapy-resistant melanoma have reported some initial findings. In this review, we summarize important mechanisms that drive resistance to immunotherapies in patients with cutaneous melanoma. We have focused on tumor intrinsic characteristics of resistance, altered immune function, and systemic factors that contribute to immunotherapy resistance in melanoma. Exploring these pathways will hopefully yield novel strategies to prevent acquired resistance and overcome existing resistance to immunotherapy treatment in patients with cutaneous melanoma.
Collapse
Affiliation(s)
- Jessica Thornton
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Chandra K Singh
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | | | - Carl A Shirley
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, United States.,William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| |
Collapse
|
363
|
Zhao J, Luo Z. Discovery of Raf Family Is a Milestone in Deciphering the Ras-Mediated Intracellular Signaling Pathway. Int J Mol Sci 2022; 23:ijms23095158. [PMID: 35563547 PMCID: PMC9101324 DOI: 10.3390/ijms23095158] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
The Ras-Raf-MEK-ERK signaling pathway, the first well-established MAPK pathway, plays essential roles in cell proliferation, survival, differentiation and development. It is activated in over 40% of human cancers owing to mutations of Ras, membrane receptor tyrosine kinases and other oncogenes. The Raf family consists of three isoforms, A-Raf, B-Raf and C-Raf. Since the first discovery of a truncated mutant of C-Raf as a transforming oncogene carried by a murine retrovirus, forty years of extensive studies have provided a wealth of information on the mechanisms underlying the activation, regulation and biological functions of the Raf family. However, the mechanisms by which activation of A-Raf and C-Raf is accomplished are still not completely understood. In contrast, B-Raf can be easily activated by binding of Ras-GTP, followed by cis-autophosphorylation of the activation loop, which accounts for the fact that this isoform is frequently mutated in many cancers, especially melanoma. The identification of oncogenic B-Raf mutations has led to accelerated drug development that targets Raf signaling in cancer. However, the effort has not proved as effective as anticipated, inasmuch as the mechanism of Raf activation involves multiple steps, factors and phosphorylation of different sites, as well as complex interactions between Raf isoforms. In this review, we will focus on the physiological complexity of the regulation of Raf kinases and their connection to the ERK phosphorylation cascade and then discuss the role of Raf in tumorigenesis and the clinical application of Raf inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Jingtong Zhao
- Queen Mary School, Nanchang University, Nanchang 330031, China;
| | - Zhijun Luo
- Queen Mary School, Nanchang University, Nanchang 330031, China;
- Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang 330031, China
- NCU-QMUL Joint Research Institute of Precision Medical Science, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
364
|
Abstract
Melanoma is the most common cause of skin cancer-related death in the United States. Cutaneous melanoma is most prevalent in the head and neck. The long-term prognosis has been poor and chemotherapy is not curative. Complete surgical resection with locally advanced disease can be challenging and melanoma is resistant to radiation. Advances made in immunotherapy and genomically targeted therapy have transformed the treatment of metastatic melanoma; as of 2021, the 5-year survival for metastatic melanoma is greater than 50%. Ongoing clinical studies are underway to integrate these life-saving therapies into the presurgical or postsurgical settings. This article reviews that effort.
Collapse
Affiliation(s)
- Jay Ponto
- Earle A. Chiles Research Institute in the Robert W. Franz Cancer Center, Providence Cancer Institute, 4805 NE Glisan Street Suite 2N35, Portland, OR 97213, USA
| | - R Bryan Bell
- Earle A. Chiles Research Institute in the Robert W. Franz Cancer Center, Providence Cancer Institute, 4805 NE Glisan Street Suite 2N35, Portland, OR 97213, USA.
| |
Collapse
|
365
|
The Use and Technique of Sentinel Node Biopsy for Skin Cancer. Plast Reconstr Surg 2022; 149:995e-1008e. [PMID: 35472052 DOI: 10.1097/prs.0000000000009010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
LEARNING OBJECTIVES After studying this article, the participant should be able to: 1. Understand the indications for and prognostic value of sentinel lymph node biopsy in skin cancer. 2. Learn the advantages and disadvantages of various modalities used alone or in combination when performing sentinel lymph node biopsy. 3. Understand how to perform sentinel lymph node biopsy in skin cancer patients. SUMMARY Advances in technique used to perform sentinel lymph node biopsy to assess lymph node status have led to increased accuracy of the procedure and improved patient outcomes.
Collapse
|
366
|
Youn S, Eurich DT, McCall M, Walker J, Smylie M, Sawyer MB. Skeletal muscle is prognostic in resected stage III malignant melanoma. Clin Nutr 2022; 41:1066-1072. [PMID: 35397311 DOI: 10.1016/j.clnu.2022.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Sarcopenia (low skeletal muscle index, SMI) and myosteatosis (low skeletal muscle radiodensity, SMD) have been associated with worse survival in cancer. This study evaluated associations of body composition with survival in patients with resected stage III melanoma. METHODS A retrospective review was performed of resected stage III melanoma patients in Alberta, Canada from 2007 to 2017. Preoperative CT scans were analyzed to determine SMI and SMD. Cohort-specific SMI and SMD cut-offs that optimally predicted overall survival (OS) were identified through stratification, in addition to testing cut-offs previously established in the literature. Overall (OS), melanoma-specific (MSS), and recurrence-free survival (RFS) were determined from date of surgery and analysed using multivariable Cox regressions with age, sex, BMI, stage subgroup, ECOG PS, and tumor location as covariates. RESULTS We included 330 patients in the final analysis. Mean age was 56 years and 62.4% of patients were male. At time of censoring 150 patients (45.6%) had died. Sarcopenia based on literature cut-offs was associated with decreased OS (HR 1.55, 95% CI 1.00-2.21, p = 0.016). Using cohort-specific cut-offs, sarcopenic patients also had significantly decreased OS (HR 1.87, 95% CI 1.27-2.76, p = 0.002). Myosteatosis defined using cohort-specific cut-offs predicted worse OS (HR 2.15, 95% CI 1.42-3.25, p < 0.001), MSS (HR 2.29, 95% CI 1.40-3.75, p = 0.001) and RFS (HR 1.52, 95% CI 1.02-2.27, p = 0.041). Increased BMI ( ≥ 25) and visceral fat index were not significantly associated with survival. CONCLUSIONS Sarcopenia and myosteatosis, defined using two sets of cut-offs, are associated with decreased OS and MSS in resected stage III melanoma.
Collapse
Affiliation(s)
- Susie Youn
- Department of Surgery, University of Alberta Hospital, 8440-112 Street, Edmonton, AB, T6G 2B7, Canada.
| | - Dean T Eurich
- University of Alberta School of Public Health, 3-300 Edmonton Clinic Health Academy, 11405-87 Ave, Edmonton, Alberta, T6G 1C9, Canada
| | - Michael McCall
- Department of Surgery, University of Alberta Hospital, 8440-112 Street, Edmonton, AB, T6G 2B7, Canada
| | - John Walker
- Cross Cancer Institute, 11560 University Ave, Edmonton, AB, T6G 1Z2, Canada
| | - Michael Smylie
- Cross Cancer Institute, 11560 University Ave, Edmonton, AB, T6G 1Z2, Canada
| | - Michael B Sawyer
- Cross Cancer Institute, 11560 University Ave, Edmonton, AB, T6G 1Z2, Canada
| |
Collapse
|
367
|
van Akkooi ACJ, Zijlker LP, Wouters MWJM. Neoadjuvant Immune Checkpoint Inhibitor Therapy in Melanoma: Efficacy, Safety and Timing. BioDrugs 2022; 36:373-380. [PMID: 35397089 PMCID: PMC9148869 DOI: 10.1007/s40259-022-00525-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 11/27/2022]
Abstract
The introduction of effective systemic therapies has significantly changed the treatment of stage III and IV melanoma. Both immune checkpoint inhibitors and targeted therapies have improved recurrence-free survival in the adjuvant setting. Recent interest has sparked for neoadjuvant systemic therapy with immune checkpoint inhibitors. The intended benefit of pre-operative treatment with immunotherapy is amongst others to enable tailoring of the surgery and adjuvant systemic therapy according to the treatment response. Most importantly, recurrence-free survival might be improved by neoadjuvant systemic therapy over the current standard of care of surgery followed by adjuvant systemic therapy. The first phase I and II trials investigating anti-PD1 inhibitors, both as a single agent and in combination with anti-CTLA-4 inhibitors or other therapeutic agents, have shown promising results. Pathological complete response on neoadjuvant systemic therapy seems a valid surrogate endpoint for relapse-free and overall survival. Pathological complete response rates in these trials vary between 30 and 70%. The optimal dose with respect to efficacy and toxicity and the interval between systemic and surgical treatment remain important issues to address. Accumulating follow-up data and ongoing phase III studies must prove if neoadjuvant systemic therapy is superior to surgery followed by standard-of-care adjuvant therapy.
Collapse
Affiliation(s)
- Alexander C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Crows Nest, PO Box 1479, Sydney, NSW, 1585, Australia.
- Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | | | - Michel W J M Wouters
- Netherlands Cancer Institute, Amsterdam, The Netherlands
- Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
368
|
Arkenau H, Taylor D, Xu X, Chitnis S, Llacer‐Perez C, Moore K, Nidamarthy PK, Ilankumaran P, De Vos‐Geelen J. Pharmacokinetic Interaction Between the MEK1/MEK2 Inhibitor Trametinib and Oral Contraceptives Containing Norethindrone and Ethinyl Estradiol in Female Patients With Solid Tumors. Clin Pharmacol Drug Dev 2022; 11:585-596. [PMID: 35157784 PMCID: PMC9304124 DOI: 10.1002/cpdd.1052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/27/2021] [Indexed: 11/12/2022]
Abstract
This phase 1 postapproval study assessed the effect of the mitogen-activated protein kinase kinase enzyme 1/enzyme 2 inhibitor trametinib (2 mg once daily, repeat dosing) on the pharmacokinetics of combined oral contraceptives (COCs) containing norethindrone (NE; 1 mg daily) and ethinyl estradiol (EE; 0.035 mg daily) in 19 female patients with solid tumors. Compared with NE/EE administered without trametinib, NE/EE administered with steady-state trametinib was associated with a clinically nonrelevant 20% increase in NE exposure (area under the curve [AUC]) and no effect on EE exposure (geometric mean ratio [geo-mean] of NE/EE + trametinib to NE/EE [90%CI]: NE AUC calculated to the end of a dosing interval at steady-state [AUCtau ] 1.20 [1.02-1.41]; NE AUC from time zero to the last measurable concentration sampling time [AUClast ] 1.2 [0.999-1.45]; EE AUCtau 1.06 [0.923-1.22]; EE AUClast 1.05 [0.883-1.25]). Maximum serum concentration (Cmax ) of NE increased by 13% and Cmax of EE decreased by 8.5% when dosed with steady-state trametinib compared with COCs administered alone (geo-mean ratio [90%CI]: NE Cmax 1.13 [0.933-1.36]; EE Cmax 0.915 [0.803-1.04]). These results indicate that repeat-dose trametinib does not lower exposure to NE or EE and, hence, is unlikely to impact the contraceptive efficacy of COCs. The pharmacokinetic parameters of trametinib and its metabolite M5 were consistent with historic data of trametinib alone. Coadministration of trametinib and COCs was generally well tolerated in this study, with observed safety signals consistent with the known safety profile of trametinib and no new reported safety events. Overall, the findings indicate that hormonal COCs can be coadministered in female patients who receive trametinib monotherapy without compromising the contraceptive efficacy.
Collapse
Affiliation(s)
- Hendrik‐Tobias Arkenau
- Sarah Cannon Research InstituteLondonUK
- Cancer InstituteUniversity College LondonLondonUK
| | | | - Xiaoying Xu
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Shripad Chitnis
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | | | - Kathleen Moore
- Stephenson Cancer CentreUniversity of OklahomaOklahoma CityOklahomaUSA
- Sarah Cannon Research InstituteNashvilleTennesseeUSA
| | | | | | - Judith De Vos‐Geelen
- Department of Internal MedicineDivision of Medical OncologyGROW, School for Oncology and Developmental BiologyMaastricht UMC+MaastrichtThe Netherlands
| |
Collapse
|
369
|
Boland G. Management of Patients With Advanced Melanoma. J Natl Compr Canc Netw 2022. [DOI: 10.6004/jnccn.2022.5003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
For treatment of melanoma, accumulated research has allowed the transition of the most effective treatments into earlier stages of disease management. To this end, immunotherapy has become approved for high-risk stage II and resected stage III melanoma, and is currently being evaluated in the neoadjuvant setting. BRAF/MEK-targeted therapy is also approved in the adjuvant setting and is being evaluated in neoadjuvant trials. In stage IV disease, the optimal sequencing of these 2 main approaches is with immunotherapy initiated first. Recently, the LAG-3 antibody relatlimab, in combination with nivolumab, has produced impressive responses with low toxicity and has become a new standard of care compared with anti–PD-1 monotherapy. As heterogeneity within disease stages has become better appreciated, staging and risk classification have been refined.
Collapse
|
370
|
Dieng M, Lord SJ, Turner RM, Nieweg OE, Menzies AM, Saw RPM, Einstein AJ, Emmett L, Thompson JF, Lo SN, Morton RL. The Impact of Surveillance Imaging Frequency on the Detection of Distant Disease for Patients with Resected Stage III Melanoma. Ann Surg Oncol 2022; 29:2871-2881. [PMID: 35142966 PMCID: PMC8990943 DOI: 10.1245/s10434-021-11231-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND It is not known whether there is a survival benefit associated with more frequent surveillance imaging in patients with resected American Joint Committee on Cancer stage III melanoma. OBJECTIVE The aim of this study was to investigate distant disease-free survival (DDFS), melanoma-specific survival (MSS), post distant recurrence MSS (dMSS), and overall survival for patients with resected stage III melanoma undergoing regular computed tomography (CT) or positron emission tomography (PET)/CT surveillance imaging at different intervals. PATIENTS AND METHODS A closely followed longitudinal cohort of patients with resected stage IIIA-D disease treated at a tertiary referral center underwent 3- to 4-monthly, 6-monthly, or 12-monthly surveillance imaging between 2000 and 2017. Survival outcomes were estimated using the Kaplan-Meier method, and log-rank tests assessed the significance of survival differences between imaging frequency groups. RESULTS Of 473 patients (IIIA, 19%; IIIB, 31%; IIIC, 49%; IIID, 1%) 30% underwent 3- to 4-monthly imaging, 10% underwent 6-monthly imaging, and 60% underwent 12-monthly imaging. After a median follow-up of 6.2 years, distant recurrence was recorded in 252 patients (53%), with 40% detected by surveillance CT or PET/CT, 43% detected clinically, and 17% with another imaging modality. Median DDFS was 5.1 years (95% confidence interval 3.9-6.6). Among 139 IIIC patients who developed distant disease, the median dMSS was 4.4 months shorter in those who underwent 3- to 4-monthly imaging than those who underwent 12-monthly imaging. CONCLUSION Selecting patients at higher risk of distant recurrence for more frequent surveillance imaging yields a higher proportion of imaging-detected distant recurrences but is not associated with improved survival. A randomized comparison of low versus high frequency imaging is needed.
Collapse
Affiliation(s)
- Mbathio Dieng
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Robin M Turner
- Centre for Biostatistics, University of Otago, Dunedin, New Zealand
| | - Omgo E Nieweg
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital and Mater Hospitals, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital and Mater Hospitals, Sydney, NSW, Australia
| | - Andrew J Einstein
- Seymour, Paul, and Gloria Milstein Division of Cardiology, Department of Medicine, and Department of Radiology, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital Sydney, Sydney, NSW, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital and Mater Hospitals, Sydney, NSW, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Rachael L Morton
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
371
|
Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, de la Cruz Merino L, Khattak MA, Schadendorf D, Long GV, Ascierto PA, Mandala M, De Galitiis F, Haydon A, Dummer R, Grob JJ, Robert C, Carlino MS, Mohr P, Poklepovic A, Sondak VK, Scolyer RA, Kirkwood JM, Chen K, Diede SJ, Ahsan S, Ibrahim N, Eggermont AMM. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet 2022; 399:1718-1729. [PMID: 35367007 DOI: 10.1016/s0140-6736(22)00562-1] [Citation(s) in RCA: 325] [Impact Index Per Article: 108.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pembrolizumab prolongs progression-free and overall survival among patients with advanced melanoma and recurrence-free survival in resected stage III disease. KEYNOTE-716 assessed pembrolizumab as adjuvant therapy in patients with completely resected, high-risk, stage II melanoma. We report results from the planned first and second interim analyses for recurrence-free survival. METHODS In this double-blind, randomised, placebo-controlled phase 3 study, involving 160 academic medical centres and hospitals in 16 countries (Australia, Belgium, Brazil, Canada, Chile, France, Germany, Israel, Italy, Japan, Poland, South Africa, Spain, Switzerland, the UK, and the USA), patients aged 12 years or older with newly diagnosed, completely resected stage IIB or IIC melanoma (TNM stage T3b or T4 with a negative sentinel lymph node biopsy) were recruited. Eligible patients were randomly assigned (1:1), in blocks of four and stratified by T-category (3b, 4a, and 4b) and paediatric status (age 12-17 years vs ≥18 years), using an interactive response technology system to intravenous pembrolizumab 200 mg (2 mg/kg in paediatric patients) or placebo every 3 weeks for 17 cycles or until disease recurrence or unacceptable toxicity. All patients, clinical investigators, and analysts were masked to treatment assignment. The primary endpoint was investigator-assessed recurrence-free survival (defined as time from randomisation to recurrence or death) in the intention-to-treat (ITT) population (ie, all patients randomly assigned to treatment). The primary endpoint was met if recurrence-free survival was significantly improved for pembrolizumab versus placebo at either the first interim analysis (after approximately 128 patients had events) or second interim analysis (after 179 patients had events) under multiplicity control. Safety was assessed in all patients randomly assigned to treatment who received at least one dose of study treatment. This study is registered with ClinicalTrials.gov, NCT03553836, and is closed to accrual. FINDINGS Between Sept 23, 2018, and Nov 4, 2020, 1182 patients were screened, of whom 976 were randomly assigned to pembrolizumab (n=487) or placebo (n=489; ITT population). The median age was 61 years (IQR 52-69) and 387 (40%) patients were female and 589 (60%) were male. 874 (90%) of 976 patients were White and 799 (82%) were not Hispanic or Latino. 483 (99%) of 487 patients in the pembrolizumab group and 486 (99%) of 489 in the placebo group received assigned treatment. At the first interim analysis (data cutoff on Dec 4, 2020; median follow-up of 14·4 months [IQR 10·2-18·7] in the pembrolizumab group and 14·3 months [10·1-18·7] in the placebo group), 54 (11%) of 487 patients in the pembrolizumab group and 82 (17%) of 489 in the placebo group had a first recurrence of disease or died (hazard ratio [HR] 0·65 [95% CI 0·46-0·92]; p=0·0066). At the second interim analysis (data cutoff on June 21, 2021; median follow-up of 20·9 months [16·7-25·3] in the pembrolizumab group and 20·9 months [16·6-25·3] in the placebo group), 72 (15%) patients in the pembrolizumab group and 115 (24%) in the placebo group had a first recurrence or died (HR 0·61 [95% CI 0·45-0·82]). Median recurrence-free survival was not reached in either group at either assessment timepoint. At the first interim analysis, grade 3-4 treatment-related adverse events occurred in 78 (16%) of 483 patients in the pembrolizumab groups versus 21 (4%) of 486 in the placebo group. At the first interim analysis, four patients died from an adverse event, all in the placebo group (one each due to pneumonia, COVID-19-related pneumonia, suicide, and recurrent cancer), and at the second interim analysis, one additional patient, who was in the pembrolizumab group, died from an adverse event (COVID-19-related pneumonia). No deaths due to study treatment occurred. INTERPRETATION Pembrolizumab as adjuvant therapy for up to approximately 1 year for stage IIB or IIC melanoma resulted in a significant reduction in the risk of disease recurrence or death versus placebo, with a manageable safety profile. FUNDING Merck Sharp & Dohme, a subsidiary of Merck & Co, Kenilworth, NJ, USA.
Collapse
Affiliation(s)
- Jason J Luke
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paola Queirolo
- Istituto Europeo di Oncologia - IRCCS, Milano; Ospedale San Martino IRCCS, Genova, Italy
| | | | - Jacek Mackiewicz
- Poznan University of Medical Sciences, Poznan, Poland; Greater Poland Cancer Center, Poznan, Poland
| | | | - Luis de la Cruz Merino
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Sevilla, Spain; Medicine Department, Universidad de Sevilla, Sevilla, Spain
| | | | - Dirk Schadendorf
- University Hospital Essen and German Cancer Consortium Partner Site, Essen, Germany
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Center, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Mario Mandala
- University of Perugia, Perugia, Italy; Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | | | - Andrew Haydon
- Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Reinhard Dummer
- University Hospital Zürich, University of Zurich, Skin Cancer Center, Zurich, Switzerland
| | | | - Caroline Robert
- Institut Gustave Roussy, Villejuif, France; Paris-Saclay University, Villejuif, France
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Westmead and Blacktown Hospitals, Sydney, NSW, Australia
| | - Peter Mohr
- Elbe Kliniken Buxtehude, Buxtehude, Germany
| | | | - Vernon K Sondak
- H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - 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 Center, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital Sydney, NSW, Australia; NSW Health Pathology, Sydney, NSW, Australia
| | | | - Ke Chen
- Merck & Co, Inc., Kenilworth, NJ, USA
| | | | | | | | - Alexander M M Eggermont
- University Medical Center Utrecht, Utrecht, Netherlands; Princess Máxima Center, Utrecht, Netherlands; Comprehensive Cancer Center Munich, Munich, Germany
| |
Collapse
|
372
|
Ma EZ, Terhune JH, Zafari Z, Blackburn KW, Olson JA, Mullins CD, Hu Y. Treat Now or Treat Later: Comparative Effectiveness of Adjuvant Therapy in Resected Stage IIIA Melanoma. J Am Coll Surg 2022; 234:521-528. [PMID: 35290271 DOI: 10.1097/xcs.0000000000000088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Adjuvant therapy for most sentinel-node-positive (stage IIIA) melanoma may have limited clinical benefit for older patients given the competing risk of non-cancer death. The objective of this study is to model the clinical effect and cost of adjuvant therapy in stage IIIA melanoma across age groups. STUDY DESIGN A Markov decision analysis model simulated the overall survival of patients with resected stage IIIA melanoma treated with adjuvant therapy vs observation. In the adjuvant approach, patients are modeled to receive adjuvant pembrolizumab (BRAF wild type) or dabrafenib/trametinib (BRAF mutant). In the observation approach, treatment is deferred until recurrence. Transition variables were derived from landmark randomized trials in adjuvant and salvage therapy. The model was analyzed for age groups spanning 40 to 89 years. The primary outcome was the number needed to treat (NNT) to prevent one melanoma-related death at 10 years. Cost per mortality avoided was estimated using Medicare reimbursement rates. RESULTS Projections for NNT among BRAF wild type patients increased by age from 14.71 (age 40 to 44) to 142.86 (age 85 to 89), with patients in cohorts over the age of 75 having an NNT over 25. The cost per mortality avoided ranged from $2.75 million (M) (age 40 to 44) to $27.57M (age 85 to 89). Corresponding values for BRAF mutant patients were as follows: NNT 18.18 to 333.33; cost per mortality avoided ranged from $2.75M to $54.70M. CONCLUSION Universal adjuvant therapy for stage IIIA melanoma is costly and provides limited clinical benefit in patients older than 75 years.
Collapse
Affiliation(s)
- Emily Z Ma
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| | - Julia H Terhune
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| | - Zafar Zafari
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| | - Kyle W Blackburn
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| | - John A Olson
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| | - C Daniel Mullins
- Department of Pharmaceutical Health Services Research (Mullins), University of Maryland Medical Center, Baltimore, MD
| | - Yinin Hu
- Department of Surgery/Division of General and Oncologic Surgery (Ma, Terhune, Zafari, Blackburn, Olson, Hu), University of Maryland Medical Center, Baltimore, MD
| |
Collapse
|
373
|
Nenclares P, Harrington KJ. Management of Head and Neck Mucosal Melanoma. Oral Maxillofac Surg Clin North Am 2022; 34:299-314. [DOI: 10.1016/j.coms.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
374
|
Hill MV, Vidri RJ, Deng M, Handorf E, Olszanski AJ, Farma JM. Real-world frequency of BRAF testing and utilization of therapies in patients with advanced melanoma. Melanoma Res 2022; 32:79-87. [PMID: 35254330 DOI: 10.1097/cmr.0000000000000795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Both BRAF/MEK targeted agents and immunotherapy are approved for the treatment of advanced melanoma. BRAF testing is recommended at the time of advanced melanoma diagnosis. In addition, little is known regarding the treatment trends for patients with BRAF mutated tumors. This investigation aims to assess the real-world prevalence of molecular testing and treatment trends for patients with BRAF mutated tumors. Using a de-identified database, patients of age ≥18 years with advanced melanoma from 2013 to 2018 were examined. Molecular testing performed within 3 months of advanced diagnosis was considered to have the test performed at the time of diagnosis. Test prevalence was calculated and compared in groups stratified by the patient, tumor and treatment factors. In total 4459 patients were included; 1936 (43.4%) stage III, 1191 (26.7%) stage IV and 1332 (29.9%) recurrent. Totally 50.4% of patients received systemic treatment; 76.4% stage IV, 71% recurrent patients and 26.7% stage III patients. However, 73.5% received first-line immunotherapy. In total 73.8% of patients had molecular testing, and 50.5% had tested at the time of advanced diagnosis. Of those tested 42% had a BRAF mutated tumor. In total 48% of these patients received first-line immunotherapy whereas 43% received a BRAF inhibitor, with increasing immunotherapy use seen over time. The majority of patients with advanced melanoma undergo molecular testing at the time of advanced diagnosis. Immunotherapy is the most commonly prescribed treatment regardless of BRAF mutational status. These results provide real-world data on the frequency of molecular testing and treatment trends for patients with advanced melanoma.
Collapse
Affiliation(s)
- Maureen V Hill
- Department of Surgical Oncology - Valley Health, Winchester, Virginia
| | | | | | | | - Anthony J Olszanski
- Department of Medical Oncology - Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
375
|
Zhou J, Wu Z, Zhang Z, Goss L, McFarland J, Nagaraja A, Xie Y, Gu S, Peng K, Zeng Y, Zhang X, Long H, Nakagawa H, Rustgi A, Diehl JA, Meyerson M, Wong KK, Bass A. Pan-ERBB kinase inhibition augments CDK4/6 inhibitor efficacy in oesophageal squamous cell carcinoma. Gut 2022; 71:665-675. [PMID: 33789967 PMCID: PMC8921580 DOI: 10.1136/gutjnl-2020-323276] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Oesophageal squamous cell carcinoma (OSCC), like other squamous carcinomas, harbour highly recurrent cell cycle pathway alterations, especially hyperactivation of the CCND1/CDK4/6 axis, raising the potential for use of existing CDK4/6 inhibitors in these cancers. Although CDK4/6 inhibition has shown striking success when combined with endocrine therapy in oestrogen receptor positive breast cancer, CDK4/6 inhibitor palbociclib monotherapy has not revealed evidence of efficacy to date in OSCC clinical studies. Herein, we sought to elucidate the identification of key dependencies in OSCC as a foundation for the selection of targets whose blockade could be combined with CDK4/6 inhibition. DESIGN We combined large-scale genomic dependency and pharmaceutical screening datasets with preclinical cell line models, to identified potential combination therapies in squamous cell cancer. RESULTS We identified sensitivity to inhibitors to the ERBB family of receptor kinases, results clearly extending beyond the previously described minority of tumours with EGFR amplification/dependence, specifically finding a subset of OSCCs with dual dependence on ERBB3 and ERBB2. Subsequently. we demonstrated marked efficacy of combined pan-ERBB and CDK4/6 inhibition in vitro and in vivo. Furthermore, we demonstrated that squamous lineage transcription factor KLF5 facilitated activation of ERBBs in OSCC. CONCLUSION These results provide clear rationale for development of combined ERBB and CDK4/6 inhibition in these cancers and raises the potential for KLF5 expression as a candidate biomarker to guide the use of these agents. These data suggested that by combining existing Food and Drug Administration (FDA)-approved agents, we have the capacity to improve therapy for OSCC and other squamous cancer.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Surgery, West China Hospital, Sichuan University, Chendu, Sichuan Province, China
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Zhong Wu
- Department of Surgery, West China Hospital, Sichuan University, Chendu, Sichuan Province, China
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Zhouwei Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Louisa Goss
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - James McFarland
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Ankur Nagaraja
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yingtian Xie
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shengqing Gu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H.Chan School of Public Health, Boston, MA, USA
| | - Ke Peng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yong Zeng
- Department of Surgery, West China Hospital, Sichuan University, Chendu, Sichuan Province, China
| | - Xiaoyang Zhang
- Department of Oncologic Sciences, Huntsman Cancer Institute; University of Utah, Salt Lake City, Utah, USA
| | - Henry Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Anil Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - J Alan Diehl
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Kwok-Kin Wong
- Division of Hematology and Medical Oncology, New York University Medical Center, New York, New York, USA
| | - Adam Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
376
|
Ernst M, Giubellino A. The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma. Biomedicines 2022; 10:822. [PMID: 35453572 PMCID: PMC9029866 DOI: 10.3390/biomedicines10040822] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant melanoma is the leading cause of death among cutaneous malignancies. While its incidence is increasing, the most recent cancer statistics show a small but clear decrease in mortality rate. This trend reflects the introduction of novel and more effective therapeutic regimens, including the two cornerstones of melanoma therapy: immunotherapies and targeted therapies. Immunotherapies exploit the highly immunogenic nature of melanoma by modulating and priming the patient's own immune system to attack the tumor. Treatments combining immunotherapies with targeted therapies, which disable the carcinogenic products of mutated cancer cells, have further increased treatment efficacy and durability. Toxicity and resistance, however, remain critical challenges to the field. The present review summarizes past treatments and novel therapeutic interventions and discusses current clinical trials and future directions.
Collapse
Affiliation(s)
| | - Alessio Giubellino
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
377
|
RAS activation induces synthetic lethality of MEK inhibition with mitochondrial oxidative metabolism in acute myeloid leukemia. Leukemia 2022; 36:1237-1252. [PMID: 35354920 PMCID: PMC9061298 DOI: 10.1038/s41375-022-01541-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Despite recent advances in acute myeloid leukemia (AML) molecular characterization and targeted therapies, a majority of AML cases still lack therapeutically actionable targets. In 127 AML cases with unmet therapeutic needs, as defined by the exclusion of ELN favorable cases and of FLT3-ITD mutations, we identified 51 (40%) cases with alterations in RAS pathway genes (RAS+, mostly NF1, NRAS, KRAS, and PTPN11 genes). In 79 homogeneously treated AML patients from this cohort, RAS+ status were associated with higher white blood cell count, higher LDH, and reduced survival. In AML models of oncogenic addiction to RAS-MEK signaling, the MEK inhibitor trametinib demonstrated antileukemic activity in vitro and in vivo. However, the efficacy of trametinib was heterogeneous in ex vivo cultures of primary RAS+ AML patient specimens. From repurposing drug screens in RAS-activated AML cells, we identified pyrvinium pamoate, an anti-helminthic agent efficiently inhibiting the growth of RAS+ primary AML cells ex vivo, preferentially in trametinib-resistant PTPN11- or KRAS-mutated samples. Metabolic and genetic complementarity between trametinib and pyrvinium pamoate translated into anti-AML synergy in vitro. Moreover, this combination inhibited the propagation of RA+ AML cells in vivo in mice, indicating a potential for future clinical development of this strategy in AML.
Collapse
|
378
|
Follow-up of primary melanoma patients with high risk of recurrence: recommendations based on evidence and consensus. Clin Transl Oncol 2022; 24:1515-1523. [PMID: 35349041 DOI: 10.1007/s12094-022-02822-x] [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: 10/15/2021] [Accepted: 02/27/2022] [Indexed: 10/18/2022]
Abstract
In spite of the good prognosis of patients with early-stage melanoma, there is a substantial proportion of them that develop local or distant relapses. With the introduction of targeted and immune therapies for advanced melanoma, including at the adjuvant setting, early detection of recurrent melanoma and/or second primary lesions is crucial to improve clinical outcomes. However, there is a lack of universal guidelines regarding both frequency of surveillance visits and diagnostic imaging and/or laboratory evaluations. In this article, a multidisciplinary expert panel recommends, after careful review of relevant data in the field, a consensus- and experience-based follow-up strategy for melanoma patients, taking into account prognostic factors and biomarkers and the high-risk periods and patterns of recurrence in each (sub) stage of the disease. Apart from the surveillance intensity, healthcare professionals should focus on patients' education to perform regular self-examinations of the skin and palpation of lymph nodes.
Collapse
|
379
|
Cassell O, Harwood C, Hovey M, Payne M. Implications of systemic adjuvant melanoma treatments for U.K. melanoma services: Results of the U.K. Melanoma Adjuvant Pathway Survey. Eur J Cancer Care (Engl) 2022; 31:e13565. [DOI: 10.1111/ecc.13565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/19/2019] [Accepted: 01/24/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Oliver Cassell
- Oxford University Hospitals NHS Foundation Trust Oxford UK
| | - Catherine Harwood
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry Queen Mary University of London; Barts Health NHS Trust London UK
| | | | - Miranda Payne
- Oxford University Hospitals NHS Foundation Trust Oxford UK
| |
Collapse
|
380
|
Carter TJ, George C, Harwood C, Nathan P. Melanoma in pregnancy: Diagnosis and management in early-stage and advanced disease. Eur J Cancer 2022; 166:240-253. [PMID: 35325701 DOI: 10.1016/j.ejca.2022.02.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/30/2022] [Accepted: 02/18/2022] [Indexed: 12/13/2022]
Abstract
Approximately one-third of women diagnosed with melanoma are of child-bearing age. The annual incidence of melanoma has risen steadily over the last 40 years, resulting in increasing numbers of women diagnosed with melanoma both during pregnancy, and post-partum. To date, there are no formal guidelines on the management of pregnancy associated melanoma (PAM), both early stage and metastatic. This article reviews the existing literature and provides a framework for the investigation and multidisciplinary management of PAM.
Collapse
Affiliation(s)
- Thomas J Carter
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood, Middlesex, UK
| | - Christina George
- Department of Dermatology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Catherine Harwood
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, Whitechapel, London, UK
| | - Paul Nathan
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood, Middlesex, UK.
| |
Collapse
|
381
|
Comparison of Two Rapid Assays for the Detection of BRAF V600 Mutations in Metastatic Melanoma including Positive Sentinel Lymph Nodes. Diagnostics (Basel) 2022; 12:diagnostics12030751. [PMID: 35328303 PMCID: PMC8947166 DOI: 10.3390/diagnostics12030751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Testing for the BRAF mutation is mandatory for the management of patients with locally advanced or metastatic melanoma. Molecular analysis based on DNA sequencing remains the gold-standard method for the screening of the different BRAF mutations. These methods must be rapid, sensitive, and specific enough to allow optimal therapeutic management in daily practice and also to include patients in clinical trials. Here, we compared the Idylla BRAF Mutation Test and the anti-BRAF V600E (clone VE1) immunohistochemistry (IHC) in 90 melanoma samples, with a focus on a challenging cohort of 32 positive sentinel lymph nodes. The BRAF status was assessed with both methods independently of the percentage of tumor cells. The concordance rate was calculated excluding both non-contributory analyses and BRAFV600K/R/M mutants due to the specific V600E-IHC test design. The incidence of the BRAFV600E mutation was 33% with both BRAF Idylla and BRAF IHC. The agreement rate was 91% (72/79). Although the agreement rate was high, we suggest that the use of IHC is more suitable for rapid BRAF testing on sentinel lymph node biopsies when associated with a low percentage and scattered tumor cells, which gave a high risk of non-contributory analysis and/or false negative results with the IdyllaTMBRAF Mutation Test.
Collapse
|
382
|
Ritter N, Peeken L, Schultz ES, Debus D. Die Systemtherapie des malignen Melanoms. AKTUELLE DERMATOLOGIE 2022. [DOI: 10.1055/a-1700-9298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
ZusammenfassungIn den vergangenen 10 Jahren wurde die Systemtherapie des malignen Melanoms durch die Zulassung neuer Substanzen revolutioniert. In der vorliegenden Übersicht werden zunächst die aktuellen adjuvanten Therapiemöglichkeiten beschrieben, anschließend werden der Kenntnisstand zur neoadjuvanten Therapie dargestellt und schließlich die Behandlungsoptionen im inoperablen Stadium beleuchtet.
Collapse
Affiliation(s)
- Nathalie Ritter
- Klinikum Nürnberg, Hautklinik, Universitätsklinik für Dermatologie der Paracelsus Medizinischen Privatuniversität, Nürnberg
| | - Lucia Peeken
- Klinikum Nürnberg, Hautklinik, Universitätsklinik für Dermatologie der Paracelsus Medizinischen Privatuniversität, Nürnberg
| | - Erwin S. Schultz
- Klinikum Nürnberg, Hautklinik, Universitätsklinik für Dermatologie der Paracelsus Medizinischen Privatuniversität, Nürnberg
| | - Dirk Debus
- Klinikum Nürnberg, Hautklinik, Universitätsklinik für Dermatologie der Paracelsus Medizinischen Privatuniversität, Nürnberg
| |
Collapse
|
383
|
Campos JF, Besson T, Berteina-Raboin S. Review on the Synthesis and Therapeutic Potential of Pyrido[2,3-d], [3,2-d], [3,4-d] and [4,3-d]pyrimidine Derivatives. Pharmaceuticals (Basel) 2022; 15:ph15030352. [PMID: 35337149 PMCID: PMC8949896 DOI: 10.3390/ph15030352] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
The objective of this review is to list the structures composed of a pyridopyrimidine moiety which have shown a therapeutic interest or have already been approved for use as therapeutics. We consider all the synthetic protocols to prepare these pyridopyrimidine derivatives. The review is organized into four sections, successively pyrido[2,3-d]pyrimidines, pyrido[3,4-d]pyrimidines, pyrido[4,3-d]pyrimidines and pyrido[3,2-d]pyrimidines. For each compound we present the biological activity and the synthetic route reported. To produce this manuscript, the bibliographic research was done using Reaxys and Scifinder for each kind of pyridopyrimidine.
Collapse
Affiliation(s)
- Joana F. Campos
- Institut de Chimie Organique et Analytique (ICOA), Université d’Orléans CNRS, ICOA UMR 7311, BP 6759, Rue de Chartres, CEDEX 2, 45067 Orléans, France;
| | - Thierry Besson
- Université de Rouen-Normandie (UNIROUEN), INSA Rouen, CNRS, COBRA UMR 6014, 76000 Rouen, France;
| | - Sabine Berteina-Raboin
- Institut de Chimie Organique et Analytique (ICOA), Université d’Orléans CNRS, ICOA UMR 7311, BP 6759, Rue de Chartres, CEDEX 2, 45067 Orléans, France;
- Correspondence: ; Tel.: +33-238-494-856
| |
Collapse
|
384
|
Jacklin C, Tan M, Sravanam S, Harrison C. Appraisal of International Guidelines for Cutaneous Melanoma Management using the AGREE II assessment tool. JPRAS Open 2022; 31:114-122. [PMID: 35024406 PMCID: PMC8732330 DOI: 10.1016/j.jpra.2021.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The evidence base behind new melanoma treatments is rapidly accumulating. This is not necessarily reflected in current guidance. A recent UK-based expert consensus statement, published in JPRAS, has called for updates to the widely accepted 2015 National Institute for Health and Care Excellence (NICE) guideline for melanoma (NG14). We aimed to compare the quality of NG14 to all other melanoma guidelines published since. METHODS We conducted a systematic search of PubMed, Medline, and online clinical practice guideline databases to identify melanoma guidelines published between 29th July 2015 and 23rd August 2021 providing recommendations for adjuvant treatment, radiotherapy, surgical management, or follow-up care. Three authors independently assessed the quality of identified guidelines using the Appraisal of Guidelines for Research & Evaluation Instrument II (AGREE II) assessment tool, which measures six domains of guideline development. Inter-rater reliability was assessed by Kendall's coefficient of concordance (W). RESULTS Twenty-nine guidelines were included and appraised with excellent concordance (Kendall's W for overall guideline score 0.88, p<0.001). Overall, melanoma guidelines scored highly in the domains of 'Scope and purpose' and 'Clarity of presentation', but poorly in the 'Applicability' domain. The NICE guideline on melanoma (NG14) achieved the best overall scores. CONCLUSION Melanoma treatment has advanced since NG14 was published, however, the NICE melanoma guideline is of higher quality than more recent alternatives. The planned update of NG14 in 2022 is in demand.
Collapse
Affiliation(s)
- C. Jacklin
- Medical Sciences Divisional Office, University of Oxford, Level 3, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - M. Tan
- Academic Section of Vascular Surgery, Department of Surgery and Cancer, Imperial College, London
| | - S. Sravanam
- Medical Sciences Divisional Office, University of Oxford, Level 3, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - C.J. Harrison
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
385
|
Eggermont AM, Ascierto PA, Khushalani NI, Schadendorf D, Boland G, Weber J, Lewis KD, Johnson D, Rivalland G, Khattak A, Majem M, Gogas H, Long GV, Currie SL, Chien D, Tagliaferri MA, Carlino MS, Diab A. PIVOT-12: a phase III study of adjuvant bempegaldesleukin plus nivolumab in resected stage III/IV melanoma at high risk for recurrence. Future Oncol 2022; 18:903-913. [PMID: 35073733 DOI: 10.2217/fon-2021-1286] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Bempegaldesleukin (BEMPEG: NKTR-214) is an immunostimulatory IL-2 cytokine prodrug engineered to deliver a controlled, sustained and preferential IL-2 pathway signal. Nivolumab (NIVO), a PD-1 inhibitor, has been shown to prolong survival in patients with advanced melanoma and recurrence-free survival in the adjuvant setting. PIVOT-02 showed that BEMPEG plus NIVO was well-tolerated and demonstrated clinical activity as first-line therapy in metastatic melanoma. PIVOT-12 is a randomized, phase III, global, multicenter, open-label study comparing adjuvant therapy with BEMPEG plus NIVO versus NIVO alone in adult and adolescent patients with completely resected cutaneous stage III/IV melanoma at high risk of recurrence. The primary objective is to compare the efficacy, as measured by recurrence-free survival, of BEMPEG plus NIVO versus NIVO.
Collapse
Affiliation(s)
- Alexander Mm Eggermont
- Princess Máxima Center for Pediatric Oncology & University Medical Center Utrecht, Utrecht, Netherlands
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Naples, Italy
| | | | - Dirk Schadendorf
- West German Cancer Center at the University Hospital Essen, Essen, Germany
| | | | - Jeffrey Weber
- Perlmutter Cancer Center at NYU Langone Health, New York, NY 10016, USA
| | - Karl D Lewis
- University of Colorado Cancer Center, Aurora, CO 80045, USA
| | | | - Gareth Rivalland
- University of Auckland & Auckland City Hospital, Auckland, New Zealand
| | - Adnan Khattak
- Hollywood Private Hospital, Edith Cowan University, Perth, Australia
| | | | - Helen Gogas
- National & Kapodistrian University of Athens, Athens, Greece
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, & Royal North Shore & Mater Hospitals, Sydney, Australia
| | - Sue L Currie
- Nektar Therapeutics, San Francisco, CA 94158, USA
| | - David Chien
- Nektar Therapeutics, San Francisco, CA 94158, USA
| | | | - Matteo S Carlino
- Westmead & Blacktown Hospitals & Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Adi Diab
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
386
|
Glen C, Tan YY, Waterston A, Evans TRJ, Jones RJ, Petrie MC, Lang NN. Mechanistic and Clinical Overview Cardiovascular Toxicity of BRAF and MEK Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:1-18. [PMID: 35492830 PMCID: PMC9040125 DOI: 10.1016/j.jaccao.2022.01.096] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Rapidly accelerated fibrosarcoma B-type (BRAF) and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors have revolutionized melanoma treatment. Approximately half of patients with melanoma harbor a BRAF gene mutation with subsequent dysregulation of the RAF-MEK-ERK signaling pathway. Targeting this pathway with BRAF and MEK blockade results in control of cell proliferation and, in most cases, disease control. These pathways also have cardioprotective effects and are necessary for normal vascular and cardiac physiology. BRAF and MEK inhibitors are associated with adverse cardiovascular effects including hypertension, left ventricular dysfunction, venous thromboembolism, atrial arrhythmia, and electrocardiographic QT interval prolongation. These effects may be underestimated in clinical trials. Baseline cardiovascular assessment and follow-up, including serial imaging and blood pressure assessment, are essential to balance optimal anti-cancer therapy while minimizing cardiovascular side effects. In this review, an overview of BRAF/MEK inhibitor-induced cardiovascular toxicity, the mechanisms underlying these, and strategies for surveillance, prevention, and treatment of these effects are provided.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- AF, atrial fibrillation
- BRAF inhibitor
- BRAF, rapidly accelerated fibrosarcoma B-type
- CVAE, cardiovascular adverse event
- EGFR, epidermal growth factor receptor
- ERK, extracellular signal-regulated kinase
- LVSD, left ventricular systolic dysfunction
- MEK inhibitor
- MEK, mitogen-activated extracellular signal-regulated kinase
- RAF, rapidly accelerated fibrosarcoma
- VEGF, vascular endothelial growth factor
- cardio-oncology
- cardiovascular toxicity
- hypertension
- left ventricular systolic dysfunction
- melanoma
Collapse
Affiliation(s)
- Claire Glen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Yun Yi Tan
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Ashita Waterston
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Thomas R. Jeffry Evans
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert J. Jones
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark C. Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| |
Collapse
|
387
|
Atkinson V, Robert C, Grob JJ, Gogas H, Dutriaux C, Demidov L, Gupta A, Menzies AM, Ryll B, Miranda F, Banerjee H, Lau M, Del Vecchio M. Improved pyrexia-related outcomes associated with an adapted pyrexia adverse event management algorithm in patients treated with adjuvant dabrafenib plus trametinib: Primary results of COMBI-APlus. Eur J Cancer 2022; 163:79-87. [PMID: 35042070 DOI: 10.1016/j.ejca.2021.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/12/2021] [Accepted: 12/13/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND COMBI-AD demonstrated long-term benefit of adjuvant dabrafenib plus trametinib in patients with resected stage III BRAF V600E/K-mutant melanoma; however, 9% of patients permanently discontinued therapy due to pyrexia. COMBI-APlus evaluated whether an adapted pyrexia management algorithm reduces high-grade pyrexia and pyrexia-related adverse outcomes. METHODS COMBI-APlus is an open-label, phase IIIb trial evaluating an adapted pyrexia management algorithm in patients with high-risk resected stage III BRAF V600E/K-mutant melanoma treated with up to 12 months of adjuvant dabrafenib plus trametinib. Both drugs were interrupted for pyrexia (temperature ≥38°C) or the occurrence of pyrexia syndrome for suspected recurrent pyrexia. Treatment was restarted at the same dose once patients were symptom free for ≥24 h. The primary endpoint was the composite rate of grade 3/4 pyrexia, hospitalisation due to pyrexia, or permanent discontinuation due to pyrexia versus historical COMBI-AD control (20.0%; 95% confidence interval [CI], 16.3%-24.1%). RESULTS At data cutoff (5 October 2020), COMBI-APlus met its primary endpoint of significant improvement in the composite rate of pyrexia (8.0% [95% CI, 5.9%-10.6%]), with rates of 3.8% for grade 3/4 pyrexia, 4.3% for hospitalisation due to pyrexia, and 2.4% for discontinuation due to pyrexia. Estimated 12-month relapse-free survival was 91.8% (95% CI, 89.0%-93.9%). The most common adverse events were consistent with those in COMBI-AD, and 14.7% of patients permanently discontinued treatment due to adverse events. CONCLUSIONS The adapted pyrexia management algorithm appears to reduce the incidence of severe pyrexia outcomes, enables patients to manage pyrexia at home, and helps patients remain on treatment. CLINICAL TRIAL REGISTRATION NCT03551626.
Collapse
Affiliation(s)
- Victoria Atkinson
- Division of Cancer Services, Princess Alexandra Hospital, University of Queensland, 199 Ipswich Road, Woolloongabba, Queensland 4102, Australia.
| | - Caroline Robert
- Department of Medicine, Gustave Roussy, Paris-Saclay University, Villejuif, Orsay, France
| | - Jean J Grob
- Dermatology and Skin Cancer Department, Aix-Marseille University, Marseille, France
| | - Helen Gogas
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Caroline Dutriaux
- Department of Dermatology, University Hospital of Bordeaux, Bordeaux, France
| | - Lev Demidov
- Tumor Biotherapy, N. N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Avinash Gupta
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Alexander M Menzies
- Department of Medical Oncology, Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Bettina Ryll
- Medical Oncology, Melanoma Patient Network Europe, Uppsala, Sweden
| | - Flora Miranda
- Clinical Operations, Global Medical Affairs, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Hiya Banerjee
- Analytics, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Mike Lau
- Global Medical Affairs, Novartis Pharma AG, Basel, Switzerland
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
388
|
Novel treatment strategy for NRAS-mutated melanoma through a selective inhibitor of CD147/VEGFR-2 interaction. Oncogene 2022; 41:2254-2264. [PMID: 35217792 DOI: 10.1038/s41388-022-02244-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/28/2022] [Accepted: 02/11/2022] [Indexed: 11/09/2022]
Abstract
More than 70% of human NRASmut melanomas are resistant to MEK inhibitors highlighting the crucial need for efficient therapeutic strategies for these tumors. CD147, a membrane receptor, is overexpressed in most cancers including melanoma and is associated with poor prognosis. We show here that CD147i, a specific inhibitor of CD147/VEGFR-2 interaction represents a potential therapeutic strategy for NRASmut melanoma cells. It significantly inhibited the malignant properties of NRASmut melanomas ex vivo and in vivo. Importantly, NRASmut patient's-derived xenografts, which were resistant to MEKi, became sensitive when combined with CD147i leading to decreased proliferation ex vivo and tumor regression in vivo. Mechanistic studies revealed that CD147i effects were mediated through STAT3 pathway. These data bring a proof of concept on the impact of the inhibition of CD147/VEGFR-2 interaction on melanoma progression and represents a new therapeutic opportunity for NRASmut melanoma when combined with MEKi.
Collapse
|
389
|
Truong TG, Kennedy LB, Patel SP. 25 Years of Adjuvant Therapy in Melanoma: A Perspective on Current Approvals and Insights into Future Directions. Curr Oncol Rep 2022; 24:533-542. [PMID: 35192117 DOI: 10.1007/s11912-022-01232-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Thach-Giao Truong
- Kaiser Permanente Northern California, 975 Sereno Drive, Vallejo, CA, 94589, USA
| | - Lucy Boyce Kennedy
- The Cleveland Clinic Foundation, 9500 Euclid Ave CA-60, Cleveland, OH, 44195, USA
| | - Sapna P Patel
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0430, Houston, TX, 77030, USA.
| |
Collapse
|
390
|
McKay DR, Nguyen P, Wang A, Hanna TP. A population-based study of administrative data linkage to measure melanoma surgical and pathology quality. PLoS One 2022; 17:e0263713. [PMID: 35180251 PMCID: PMC8856577 DOI: 10.1371/journal.pone.0263713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/25/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Continuous quality improvement is important for cancer systems. However, collecting and compiling quality indicator data can be time-consuming and resource-intensive. Here we explore the utility and feasibility of linked routinely collected health data to capture key elements of quality of care for melanoma in a single-payer, universal health care setting.
Method
This pilot study utilized a retrospective population-based cohort from a previously developed linked administrative data set, with a 65% random sample of all invasive cutaneous melanoma cases diagnosed 2007–2012 in the province of Ontario. Data from the Ontario Cancer Registry was utilized, supplemented with linked pathology report data from Cancer Care Ontario, and other linked administrative data describing health care utilization. Quality indicators identified through provincial guidelines and international consensus were evaluated for potential collection with administrative data and measured where possible.
Results
A total of 7,654 cases of melanoma were evaluated. Ten of 25 (40%) candidate quality indicators were feasible to be collected with the available administrative data. Many indicators (8/25) could not be measured due to unavailable clinical information (e.g. width of clinical margins). Insufficient pathology information (6/25) or health structure information (1/25) were less common reasons. Reporting of recommended variables in pathology reports varied from 65.2% (satellitosis) to 99.6% (body location). For stage IB-II or T1b-T4a melanoma patients where SLNB should be discussed, approximately two-thirds met with a surgeon experienced in SLNB. Of patients undergoing full lymph node dissection, 76.2% had adequate evaluation of the basin.
Conclusions
We found that use of linked administrative data sources is feasible for measurement of melanoma quality in some cases. In those cases, findings suggest opportunities for quality improvement. Consultation with surgeons offering SLNB was limited, and pathology report completeness was sub-optimal, but was prior to routine synoptic reporting. However, to measure more quality indicators, text-based data sources will require alternative approaches to manual collection such as natural language processing or standardized collection. We recommend development of robust data platforms to support continuous re-evaluation of melanoma quality indicators, with the goal of optimizing quality of care for melanoma patients on an ongoing basis.
Collapse
Affiliation(s)
- Douglas R. McKay
- Division of Plastic Surgery, Department of Surgery, Queen’s University, Kingston, Ontario, Canada
| | - Paul Nguyen
- ICES at Queen’s University, Kingston, Ontario, Canada
| | - Ami Wang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Timothy P. Hanna
- ICES at Queen’s University, Kingston, Ontario, Canada
- Department of Oncology, Queen’s University, Kingston, Ontario, Canada
- Division of Cancer Care and Epidemiology, Cancer Research Institute at Queen’s University, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
391
|
Amabile S, Tonella L, Rubatto M, Argenziano G, Babino G, Frascione P, Dika E, Giacomelli L, Parodi A, Quaglino P. Clinical evaluation of hydration index using a corneometer in metastatic melanoma patients treated with BRAF/MEK inhibitors: a prospective study. Eur J Dermatol 2022; 32:ejd.2022.4198. [PMID: 35153191 PMCID: PMC9170549 DOI: 10.1684/ejd.2022.4198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Simone Amabile
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Luca Tonella
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Marco Rubatto
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | | | | | - Pasquale Frascione
- Oncological Dermatology and Skin Tumor Prevention Unit, Dermatological Hospital “San Gallicano”, Rome, Italy
| | - Emi Dika
- Dermatology, IRCCS of Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Dermatology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Aurora Parodi
- Dermatologic Clinic, University of Genova, Genova, Italy
| | - Pietro Quaglino
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| |
Collapse
|
392
|
Melixetian M, Pelicci PG, Lanfrancone L. Regulation of LncRNAs in Melanoma and Their Functional Roles in the Metastatic Process. Cells 2022; 11:577. [PMID: 35159386 PMCID: PMC8834033 DOI: 10.3390/cells11030577] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are key regulators of numerous intracellular processes leading to tumorigenesis. They are frequently deregulated in cancer, functioning as oncogenes or tumor suppressors. As they act through multiple mechanisms, it is not surprising that they may exert dual functions in the same tumor. In melanoma, a highly invasive and metastatic tumor with the propensity to rapidly develop drug resistance, lncRNAs play different roles in: (i) guiding the phenotype switch and leading to metastasis formation; (ii) predicting the response of melanoma patients to immunotherapy; (iii) triggering adaptive responses to therapy and acquisition of drug resistance phenotypes. In this review we summarize the most recent findings on the lncRNAs involved in melanoma growth and spreading to distant sites, focusing on their role as biomarkers for disease diagnosis and patient prognosis, or targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marine Melixetian
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| |
Collapse
|
393
|
Homan D, Ruissen-Luijt P, Boekhout A, Maaskant J. A historic cohort study of nutritional status related side effects and weight loss of cancer patients treated with immunotherapy. Clin Nutr ESPEN 2022; 47:163-169. [DOI: 10.1016/j.clnesp.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022]
|
394
|
Gouda MA, Polivka J, Huang HJ, Treskova I, Pivovarcikova K, Fikrle T, Woznica V, Dustin DJ, Call SG, Meric-Bernstam F, Pesta M, Janku F. Ultrasensitive detection of BRAF mutations in circulating tumor DNA of non-metastatic melanoma. ESMO Open 2022; 7:100357. [PMID: 34942440 PMCID: PMC8695283 DOI: 10.1016/j.esmoop.2021.100357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/21/2022] Open
Abstract
Background Implementation of adjuvant therapies in non-metastatic melanoma improved treatment outcomes in some patients; however, adjuvant therapy can be associated with significant cost and risk of toxicity. Therefore, there is an unmet need to better identify patients at high risk of recurrence. Patients and methods We carried out an ultrasensitive droplet digital PCR (ddPCR)-based detection of BRAFV600E-mutated circulating tumor DNA (ctDNA) from blood samples prospectively collected before surgery, 1 hour after surgery, and then serially during follow-up. Results In 80 patients (stages ≤III), BRAFV600E mutations were detected in 47.2% of tissue, in 37.7% of ctDNA samples collected before surgery, and in 25.9% of ctDNA samples collected 1 hour after surgery. Patients with detected ctDNA in blood collected 1 hour after surgery compared to patients without detected ctDNA had higher likelihood of melanoma recurrence (P < 0.001) and shorter median disease-free survival (P = 0.001) and overall survival (P = 0.003). Conclusions Ultrasensitive ddPCR can detect ctDNA in pre- and post-surgical blood samples from patients with resectable melanoma. Detection of ctDNA in post-surgical samples is associated with inferior treatment outcomes. Ultrasensitive ddPCR can detect ctDNA in pre- and post-surgical samples. Detection of ctDNA 1 hour after surgery is associated with inferior treatment outcomes. There were no associations between ctDNA detection at other timepoints and clinical outcomes.
Collapse
Affiliation(s)
- M A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Polivka
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Department of Neurology, University Hospital Pilsen, Pilsen, Czech Republic
| | - H J Huang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - I Treskova
- Department of Plastic Surgery, University Hospital Pilsen, Pilsen, Czech Republic
| | - K Pivovarcikova
- Department of Pathology, University Hospital Pilsen, Pilsen, Czech Republic
| | - T Fikrle
- Department of Dermatovenerology, University Hospital Pilsen, Pilsen, Czech Republic
| | - V Woznica
- Department of Plastic Surgery, University Hospital Pilsen, Pilsen, Czech Republic
| | - D J Dustin
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S G Call
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M Pesta
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Department of Biology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - F Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
395
|
Williams TS, Tallon B, Adams BM. Melanoma sentinel lymph node biopsy and completion lymph node dissection: A regional hospital experience. J Plast Reconstr Aesthet Surg 2022; 75:730-736. [PMID: 34789434 DOI: 10.1016/j.bjps.2021.09.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 11/23/2022]
Abstract
Completion lymph node dissection (CLND) following positive sentinel lymph node biopsy (SLNB) for cutaneous melanoma is a topic of controversy. The second Multicenter Selective Lymphadenectomy Trial (MSLT-II) suggested no survival benefit with CLND over observation amongst patients with a positive SLNB. The findings of the MSLT-II may have limited applicability to our high-risk population where nodal ultrasound and non-surgical melanoma treatment is rationed. In this regional, retrospective study, we reviewed primary melanoma, SLNB and CLND histopathological reports in the Bay of Plenty District Health Board (BOPDHB) across a 10-year period. The primary outcomes measured were size of sentinel lymph node metastases and non-sentinel node (NSN) positivity on CLND for patients with a positive SLNB. In the 157 SLNB identified, the mean sentinel lymph node metastatic deposit size was larger in BOPDHB compared with MSLT-II (3.53 vs 1.07/1.11mm). A greater proportion of BOPDHB patients (54.8%) had metastatic deposits larger than 1mm compared with MSLT-II (33.2/34.5%) and the rate of NSN involvement on CLND was also higher (23.8% vs 11.5%). These findings indicate that the BOPDHB is a high-risk population for nodal melanoma metastases. Forgoing CLND in the context of a positive SLNB may place these patients at risk.
Collapse
Affiliation(s)
| | - Ben Tallon
- Department of Dermatopathology, Pathlab, Tauranga, New Zealand
| | | |
Collapse
|
396
|
Morrison SL, Han G, Elenwa F, Vetto JT, Fowler G, Leong SP, Kashani-Sabet M, Pockaj BA, Kosiorek HE, Zager JS, Sondak VK, Messina JL, Mozzillo N, Schneebaum S, Han D. Is the presence of tumor-infiltrating lymphocytes predictive of outcomes in patients with melanoma? Cancer 2022; 128:1418-1428. [PMID: 35103302 DOI: 10.1002/cncr.34013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The significance of tumor-infiltrating lymphocytes (TILs) in melanoma is debated. This article presents a multicenter, retrospective study assessing the predictive and prognostic value of TILs. METHODS The Sentinel Lymph Node Working Group database was queried from 1993 to 2018 for cases with known TIL data. TILs were categorized as absent or present, which included nonbrisk (NB), brisk (B), and present but unspecified TIL levels. Clinicopathologic factors were correlated with TILs, sentinel lymph node (SLN) status, and melanoma-specific survival (MSS). RESULTS Overall, 3203 patients were included. The median thickness was 1.5 mm, and 469 cases had SLN metastases. TILs were present in 2458 cases (76.7%), with NB, B, and unspecified TILs seen in 1691 (68.8%), 691 (28.1%), and 76 (3.1%), respectively. Multivariable analysis showed that the presence of TILs significantly predicted a negative SLN biopsy (P < .05). The median follow-up was 25.2 months. MSS was significantly better for cases with TILs than cases without TILs (P < .001). According to multivariable analysis, age, gender, thickness, mitotic rate, ulceration, lymphovascular invasion, and SLN status were significantly prognostic of MSS (all P values < .05). Although TILs were not prognostic of MSS, when multiple imputation was used and the SLN status was excluded, the presence of TILs was significantly prognostic of improved MSS (hazard ratio, 0.78; 95% confidence interval, 0.64-0.95; P = .0154). CONCLUSIONS TILs are a favorable marker because their presence significantly predicts a negative SLN, and the absence of TILs may be a prognostic marker of worse survival in patients with a positive SLN but not a negative SLN. TILs may also serve as a prognostic marker of survival when the SLN status is not considered.
Collapse
Affiliation(s)
- Steven L Morrison
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas
| | - Faith Elenwa
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas
| | - John T Vetto
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Graham Fowler
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | - Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, California
| | | | | | | | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Jane L Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | | | - Dale Han
- Division of Surgical Oncology, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
397
|
Peil J, Bock F, Kiefer F, Schmidt R, Heindl LM, Cursiefen C, Schlereth SL. New Therapeutic Approaches for Conjunctival Melanoma-What We Know So Far and Where Therapy Is Potentially Heading: Focus on Lymphatic Vessels and Dendritic Cells. Int J Mol Sci 2022; 23:1478. [PMID: 35163401 PMCID: PMC8835854 DOI: 10.3390/ijms23031478] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/25/2022] Open
Abstract
Conjunctival melanoma (CM) accounts for 5% of all ocular melanomas and arises from malignantly transformed melanocytes in the conjunctival epithelium. Current therapies using surgical excision in combination with chemo- or cryotherapy still have high rates for recurrences and metastatic disease. Lately, novel signal transduction-targeted and immune checkpoint inhibitors like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, programmed cell death protein-1 (PD-1) receptor inhibitors, BRAF- or MEK-inhibitors for systemic treatment of melanoma have improved the outcome even for unresectable cutaneous melanoma, improving patient survival dramatically. The use of these therapies is now also recommended for CM; however, the immunological background of CM is barely known, underlining the need for research to better understand the immunological basics when treating CM patients with immunomodulatory therapies. Immune checkpoint inhibitors activate tumor defense by interrupting inhibitory interactions between tumor cells and T lymphocytes at the so-called checkpoints. The tumor cells exploit these inhibitory targets on T-cells that are usually used by dendritic cells (DCs). DCs are antigen-presenting cells at the forefront of immune response induction. They contribute to immune tolerance and immune defense but in the case of tumor development, immune tolerance is often prevalent. Enhancing the immune response via DCs, interfering with the lymphatic pathways during immune cell migration and tumor development and specifically targeting tumor cells is a major therapeutic opportunity for many tumor entities including CM. This review summarizes the current knowledge on the function of lymphatic vessels in tumor growth and immune cell transport and continues to compare DC subsets in CM with related melanomas, such as cutaneous melanoma and mucosal melanoma.
Collapse
Affiliation(s)
- Jennifer Peil
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.P.); (F.B.); (L.M.H.); (C.C.)
| | - Felix Bock
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.P.); (F.B.); (L.M.H.); (C.C.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
| | - Friedemann Kiefer
- European Institute for Molecular Imaging (EIMI), University of Münster, 48149 Münster, Germany;
| | - Rebecca Schmidt
- Department of Oral, Maxillofacial and Plastic Facial Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Ludwig M. Heindl
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.P.); (F.B.); (L.M.H.); (C.C.)
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.P.); (F.B.); (L.M.H.); (C.C.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
| | - Simona L. Schlereth
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (J.P.); (F.B.); (L.M.H.); (C.C.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
398
|
Wahler S, Müller A, Fuchs S, von der Schulenburg JM. Adjuvant treatment of high-risk melanoma - cost-effectiveness analysis of treatment options for BRAF 600 mutated tumors. HEALTH ECONOMICS REVIEW 2022; 12:8. [PMID: 35059911 PMCID: PMC8780795 DOI: 10.1186/s13561-021-00347-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/08/2021] [Indexed: 05/22/2023]
Abstract
INTRODUCTION Until recently, adjuvant treatment options for higher stage resectable cutaneous melanoma were limited. Two studies with a similar set-up, published 2017, led to registration of targeted therapy for BRAF-mutated melanoma with dabrafenib and trametinib as well as of the immunotherapy with nivolumab irrespective of BRAF-mutation status. Both options have been positively assessed in Germany since 2019 for the adjuvant treatment of BRAF-V600 mutated melanoma. This study evaluates the cost-effectiveness of both treatment alternatives (dabrafenib/trametinib and nivolumab) against observation as a comparative therapy from the perspective of German statutory health funds. METHODS Partitioned survival analysis based on published survival curves for the investigated treatment options was used for a cohort model for the health states relapse free survival, progression, and death. The partitioned survival analysis approach was based on the survival curves published for the key studies Combi AD and Checkmate-238. The modelling was performed for the remaining lifetime for a cohort with starting age of 50 years. For extrapolation of the survival curves, convergence to general population mortality rates was assumed in the long term. Within the progression state, a Markov model uses three levels of progressions (locoregional, distant metastases with 1st and 2nd line treatment). Lifetime treatment costs were calculated using the German statutory health fund reimbursement scheme. Quality adjusted life years (QALYs) associated to the health states were adopted from previously published utilities based on the Combi AD study. RESULTS The treatment with dabrafenib/trametinib yielded an increase in quality adjusted life years of 2.28 QALY at an incremental lifetime cost of 86.1 T€. The incremental cost effectiveness ratio of dabrafenib/trametinib and nivolumab was comparable with 37.8 T€/QALY and 30.0 T€/QALY, respectively. Several sensitivity analyses proved the result to be insensitive. General model parameters like discount rate and length of the time horizon had stronger influence. For nivolumab, the model showed lower discounted lifetime costs (118.1 T€) compared to dabrafenib/trametinib [155.1 T€], associated with a lower gain in QALYs (1.64 years) compared to observation. CONCLUSION Both dabrafenib/trametinib and nivolumab turned out to be cost effective within internationally accepted Incremental Cost Effectiveness Ratio (ICER) thresholds with comparable cost effectiveness ratios.
Collapse
Affiliation(s)
- Steffen Wahler
- St. Bernward GmbH, Friedrich-Kirsten-Straße 40, D-22391, Hamburg, Germany.
| | - Alfred Müller
- Analytic Services GmbH, Jahnstr. 34c, D-80469, Munich, Germany
| | - Sabine Fuchs
- Novartis Pharma GmbH, Roonstr. 25, D-90429, Nuremberg, Germany
| | | |
Collapse
|
399
|
Du J, Lan T, Liao H, Feng X, Chen X, Liao W, Hou G, Xu L, Feng Q, Xie K, Liao M, Chen X, Huang J, Yuan K, Zeng Y. CircNFIB inhibits tumor growth and metastasis through suppressing MEK1/ERK signaling in intrahepatic cholangiocarcinoma. Mol Cancer 2022; 21:18. [PMID: 35039066 PMCID: PMC8762882 DOI: 10.1186/s12943-021-01482-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Considerable evidence shows that circular RNAs (circRNAs) play an important role in tumor development. However, their function in intrahepatic cholangiocarcinoma (ICC) metastasis and the underlying mechanisms are incompletely understood. METHODS circNFIB (hsa_circ_0086376, termed as cNFIB hereafter) was identified in human ICC tissues through circRNAs sequencing. The biological role of cNFIB was determined in vitro and in vivo by gain or loss of functional experiments. Fluorescence in situ hybridization (FISH), RNA immunoprecipitation (RIP) and RNA pull-down assays were conducted to analyze the interaction of cNFIB with dual specificity mitogen-activated protein kinase kinase1 (MEK1). Duolink in situ proximity ligation assay (PLA) and coimmunoprecipitation (co-IP) assay were used to investigate the effects of cNFIB on the interaction between MEK1 and mitogen-activated protein kinase 2 (ERK2). Finally, a series of in vitro and in vivo experiments were performed to explore the influences of cNFIB on the anti-tumor activity of trametinib (a MEK inhibitor). RESULTS cNFIB was significantly down-regulated in human ICC tissues with postoperative metastases. The loss of cNFIB was highly associated with aggressive characteristics and predicted unfavorable prognosis in ICC patients. Functional studies revealed that cNFIB inhibited the proliferation and metastasis of ICC cells in vitro and in vivo. Mechanistically, cNFIB competitively interacted with MEK1, which induced the dissociation between MEK1 and ERK2, thereby resulting in the suppression of ERK signaling and tumor metastasis. Moreover, we found that ICC cells with high levels of cNFIB held the potential to delay the trametinib resistance. Consistently, in vivo and in vitro studies demonstrated that cotreatment with trametinib and lentivirus vector encoding cNFIB showed greater inhibitory effect than isolated trametinib treatment. CONCLUSIONS Our findings identified that cNFIB played a key role in ICC growth and metastasis by regulating MEK1/ERK signaling. Given the efficacy of cNFIB modulation on ICC suppression and trametinib sensitivity, cNFIB appears to be a potential therapeutic molecule for ICC treatment.
Collapse
Affiliation(s)
- Jinpeng Du
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Tian Lan
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Haotian Liao
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xuping Feng
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xing Chen
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Wenwei Liao
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Guimin Hou
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Lin Xu
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingbo Feng
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Kunlin Xie
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Mingheng Liao
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiwei Huang
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Kefei Yuan
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Zeng
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
400
|
Schipper LJ, Monkhorst K, Samsom KG, Bosch LJ, Snaebjornsson P, van Boven H, Roepman P, van der Kolk LE, van Houdt WJ, van der Graaf WT, Meijer GA, Voest EE. Clinical Impact of Prospective Whole Genome Sequencing in Sarcoma Patients. Cancers (Basel) 2022; 14:436. [PMID: 35053600 PMCID: PMC8773512 DOI: 10.3390/cancers14020436] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/21/2022] Open
Abstract
With more than 70 different histological sarcoma subtypes, accurate classification can be challenging. Although characteristic genetic events can largely facilitate pathological assessment, large-scale molecular profiling generally is not part of regular diagnostic workflows for sarcoma patients. We hypothesized that whole genome sequencing (WGS) optimizes clinical care of sarcoma patients by detection of diagnostic and actionable genomic characteristics, and of underlying hereditary conditions. WGS of tumor and germline DNA was incorporated in the diagnostic work-up of 83 patients with a (presumed) sarcomas in a tertiary referral center. Clinical follow-up data were collected prospectively to assess impact of WGS on clinical decision making. In 12/83 patients (14%), the genomic profile led to revision of cancer diagnosis, with change of treatment plan in eight. All twelve patients had undergone multiple tissue retrieval procedures and immunohistopathological assessments by regional and expert pathologists prior to WGS analysis. Actionable biomarkers with therapeutic potential were identified for 30/83 patients. Pathogenic germline variants were present in seven patients. In conclusion, unbiased genomic characterization with WGS identifies genomic biomarkers with direct clinical implications for sarcoma patients. Given the diagnostic complexity and high unmet need for new treatment opportunities in sarcoma patients, WGS can be an important extension of the diagnostic arsenal of pathologists.
Collapse
Affiliation(s)
- Luuk J. Schipper
- Department of Molecular Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Kim Monkhorst
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Kris G. Samsom
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Linda J.W. Bosch
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Petur Snaebjornsson
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Hester van Boven
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Paul Roepman
- Hartwig Medical Foundation, 1098 XH Amsterdam, The Netherlands;
| | - Lizet E. van der Kolk
- Family Cancer Clinic, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Winan J. van Houdt
- Department of Surgical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | | | - Gerrit A. Meijer
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (K.M.); (K.G.S.); (L.J.W.B.); (P.S.); (H.v.B.); (G.A.M.)
| | - Emile E. Voest
- Department of Molecular Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| |
Collapse
|