51
|
Ngo HX, Oh E, Li C, Yu J. Oncology Dose Selection in Subsequent Indications: What Can We Learn From FDA-approved Oncology Drugs? Clin Ther 2024; 46:927-937. [PMID: 39304367 DOI: 10.1016/j.clinthera.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE The modern oncology drug development landscape has shifted away from traditional cytotoxic chemotherapies. Following their initial approvals, many oncology drugs have been approved in subsequent indications either as monotherapy or in combination to benefit a broader patient population. To date, dose selection strategies for subsequent indications have not been systematically reviewed. This review examines how approved dosing regimens were selected in subsequent indications for FDA-approved oncology drugs. METHODS The Drugs@FDA database was used to identify FDA-approved new molecular entities (NMEs) between 2010 and 2023. NMEs with more than 1 approved indication were included in the analysis. In total, the dosing regimens for 67 novel oncology drugs that obtained FDA approvals for multiple indications were evaluated. FINDINGS Overall, in subsequent indications, 72% of NMEs used the same or clinically equivalent alternative dosing regimens to those approved in the initial indications. Amongst the 28% of NMEs that used different dosing regimens, safety/tolerability was the leading cause of a dosing regimen changes in both monotherapy and combination therapy settings. Other factors leading to changes in dosing regimens include differences in tumor biology, disease burden, pharmacokinetics, and overall benefit-risk profiles obtained from dose-finding studies. IMPLICATIONS Our analysis highlighted the importance of selecting a safe, tolerable, and yet efficacious dosing regimen for the initial indication as a suboptimal initially approved regimen could lead to dosing regimen changes in later indications. Preclinical and clinical data could be leveraged to understand the pharmacology, pharmacokinetic, and pharmacodynamic differences between indications and thus support dose selection in subsequent indications.
Collapse
Affiliation(s)
- Huy X Ngo
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Elise Oh
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Jiajie Yu
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA.
| |
Collapse
|
52
|
Woodford R, McKeown J, Hoeijmakers LL, Mangana J, Dimitriou F, Allayous C, Zaman F, Aya F, Marsiglio J, Goodman R, Rayson V, Placzke J, Kessels J, Ramalyte E, Haque W, Wilson I, Trojaniello C, Benannoune N, Roberts-Thomson R, Robert C, Blank CU, Dummer R, Lebbe C, Haydon A, Arance A, Hu-Lieskovan S, Johnson DB, Mcarthur GA, Rutkowski P, Neyns B, Sullivan RJ, Weber J, Carlino MS, Ascierto PA, Lo S, Long GV, Menzies AM. Nature and management of melanoma recurrences following adjuvant anti-PD-1 based therapy. Eur J Cancer 2024; 212:115055. [PMID: 39366209 DOI: 10.1016/j.ejca.2024.115055] [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: 07/02/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
INTRODUCTION Approximately 50 % of resected stage II-IV melanoma patients develop recurrent disease by 5 years despite adjuvant anti-PD-1 therapy. Data to define best management of recurrences is lacking. METHODS This was a multicentre, international, retrospective cohort study. Patients with resected stage II-IV melanoma who commenced adjuvant anti-PD-1-based therapy before January 2022 and later recurred were identified. Data on demographics, disease characteristics, recurrence patterns, management and outcomes were collected. RESULTS 711 patients from 17 sites were included. Median age was 60 [range 16-92], 64 % were male, 2 % stage II, 91 % were stage III, 7 % stage IV. Median time to recurrence was 6.2 months (0-68.5) and median follow up time from recurrence was 19.8 months (range 0.2-73.1). 63 % recurred on anti-PD-1 therapy, 36 % off therapy [3 % < 6 months, 33 % > 6 months]. Initial recurrences were locoregional (LR) alone in 44 %, distant alone (DR) in 43 %, and 11 % in both sites. LR recurrences were managed with local therapy, alone (62 %) or with "second adjuvant" anti-PD-1 (14 %) or BRAF/MEK therapy (23 %); 12 m RFS2 was 25 %, 29 % and 69 % respectively (p = 0.0045). Definitive systemic therapy at first recurrence was given in 16 % LR and 86 % DR, with best outcomes for anti-CTLA4 + anti-PD-1 and trial combinations (24 m PFS 63 % and 69 %, respectively). The 24 m OS for the entire cohort was 65 %. CONCLUSION Most recurrences following adjuvant anti-PD-1 based therapy occur early and while still on drug. Outcomes are poor, regardless of site, timing of recurrence, and subsequent treatment.
Collapse
Affiliation(s)
- Rachel Woodford
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Janet McKeown
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Lotte L Hoeijmakers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, Amsterdam, The Netherlands
| | - Johanna Mangana
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Clara Allayous
- APHP Hospital St-Louis, Dermatology Department, DMU ICARE, Paris, France
| | - Farzana Zaman
- Department of Medical Oncology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia
| | - Francisco Aya
- Hospital Clinic Barcelona, Calle de Villaroel 170, Barcelona, Spain
| | - John Marsiglio
- Huntsman Cancer Institute, University of Utah School of Medicine, UT, USA
| | - Rachel Goodman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Victoria Rayson
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia
| | - Joanna Placzke
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skowska-Curie Research Institute of Oncology, Masovian Voivodeship, Warsaw, Poland
| | - Jolien Kessels
- Department of Medical Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Belgium
| | | | - Waqas Haque
- Department of Medicine, New York University Grossman School of Medicine Langone, Manhattan, NY, USA
| | - Isabella Wilson
- Department of Medical Oncology, Westmead and Blacktown Hospitals, NSW, Australia
| | - Claudia Trojaniello
- Unit of Melanoma Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Naima Benannoune
- Dermatology Unit, Gustave-Roussy Paris Saclay University, 39 Rue Camille Desmoulins, Villejuif, France
| | | | - Caroline Robert
- Dermatology Unit, Gustave-Roussy Paris Saclay University, 39 Rue Camille Desmoulins, Villejuif, France
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, Amsterdam, The Netherlands
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Ramistrasse 100, Zurich, Switzerland
| | - Celeste Lebbe
- APHP Hospital St-Louis, Dermatology Department, DMU ICARE, Paris, France; Universite de Paris Cité, 45 Rue des Saint-Pères, Paris, France
| | - Andrew Haydon
- Department of Medical Oncology, The Alfred Hospital, 55 Commercial Road, Melbourne, Victoria, Australia
| | - Ana Arance
- Hospital Clinic Barcelona, Calle de Villaroel 170, Barcelona, Spain
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, UT, USA
| | | | - Grant A Mcarthur
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skowska-Curie Research Institute of Oncology, Masovian Voivodeship, Warsaw, Poland
| | - Bart Neyns
- Department of Medical Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Belgium
| | | | - Jeffrey Weber
- Department of Medicine, New York University Grossman School of Medicine Langone, Manhattan, NY, USA
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Westmead and Blacktown Hospitals, NSW, Australia
| | - Paolo A Ascierto
- Unit of Melanoma Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Serigne Lo
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, North Sydney, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, 45 Rocklands Road, Wollstonecraft, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, North Sydney, Sydney, NSW, Australia.
| |
Collapse
|
53
|
Jalil A, Donate MM, Mattei J. Exploring resistance to immune checkpoint inhibitors and targeted therapies in melanoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:42. [PMID: 39534873 PMCID: PMC11555183 DOI: 10.20517/cdr.2024.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Melanoma is the most aggressive form of skin cancer, characterized by a poor prognosis, and its incidence has risen rapidly over the past 30 years. Recent therapies, notably immunotherapy and targeted therapy, have significantly improved the outcome of patients with metastatic melanoma. Previously dismal five-year survival rates of below 5% have shifted to over 50% of patients surviving the five-year mark, marking a significant shift in the landscape of melanoma treatment and survival. Unfortunately, about 50% of patients either do not respond to therapy or experience early or late relapses following an initial response. The underlying mechanisms for primary and secondary resistance to targeted therapies or immunotherapy and relapse patterns remain not fully identified. However, several molecular pathways and genetic factors have been associated with melanoma resistance to these treatments. Understanding these mechanisms paves the way for creating novel treatments that can address resistance and ultimately enhance patient outcomes in melanoma. This review explores the mechanisms behind immunotherapy and targeted therapy resistance in melanoma patients. Additionally, it describes the treatment strategies to overcome resistance, which have improved patients' outcomes in clinical trials and practice.
Collapse
Affiliation(s)
- Anum Jalil
- Department of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Melissa M Donate
- Long School of Medicine, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| | - Jane Mattei
- Department of Hematology Oncology, UT Health Science Center San Antonio, San Antonio, TA 78229, USA
| |
Collapse
|
54
|
Babaei S, Fadaee M, Abbasi-Kenarsari H, Shanehbandi D, Kazemi T. Exosome-based immunotherapy as an innovative therapeutic approach in melanoma. Cell Commun Signal 2024; 22:527. [PMID: 39482766 PMCID: PMC11526674 DOI: 10.1186/s12964-024-01906-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
The malignant form of melanoma is one of the deadliest human cancers that accounts for almost all of the skin tumor-related fatalities in its later stages. Achieving an exhaustive understanding of reliable cancer-specific markers and molecular pathways can provide numerous practical techniques and direct the way toward the development of rational curative medicines to increase the lifespan of patients. Immunotherapy has significantly enhanced the treatment of metastatic and late-stage melanoma, resulting in an incredible increase in positive responses to therapy. Despite the increasing occurrence of melanoma, the median survival rate for patients with advanced, inoperable terminal disease has increased from around six months to almost six years. The current knowledge of the tumor microenvironment (TME) and its interaction with the immune system has resulted in the swift growth of innovative immunotherapy treatments. Exosomes are small extracellular vesicles (EVs), ranging from 30 to 150 nm in size, that the majority of cells released them. Exosomes possess natural advantages such as high compatibility with living organisms and low potential for causing immune reactions, making them practical for delivering therapeutic agents like chemotherapy drugs, nucleic acids, and proteins. This review highlights recent advancements in using exosomes as an approach to providing medications for the treatment of melanoma.
Collapse
Affiliation(s)
- Shabnam Babaei
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Abbasi-Kenarsari
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Dariush Shanehbandi
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center , Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran, P.O. Box: 5165683146.
| |
Collapse
|
55
|
Therien AD, Chime-Eze CM, Rhodin KE, Beasley GM. Neoadjuvant therapy for melanoma: past, present, and future. Surg Oncol 2024; 56:102127. [PMID: 39236515 DOI: 10.1016/j.suronc.2024.102127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
Modern systemic therapy has dramatically improved outcomes for many patients with advanced metastatic melanoma. The success of these therapies has attracted much scientific interest while these therapies have made their way into the treatment of earlier stages of disease. Randomized trials have led to the approval of adjuvant immunotherapy and targeted therapy for resected stage III melanoma. However, most recently, these therapies have gained traction in the neoadjuvant setting. Promising early results led to randomized controlled trials that have now established neoadjuvant therapy as standard of care in advanced melanoma patients. Questions remain regarding the optimal choice of therapy, duration and timing of neoadjuvant therapy, extent of surgery, and the need for additional adjuvant therapy for patients who received neoadjuvant therapy. Herein we provide an overview of neoadjuvant therapy for melanoma and dilemmas to its broader applications.
Collapse
Affiliation(s)
| | | | - Kristen E Rhodin
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
56
|
Pardessus Otero A, Rafecas-Codern A, Porcel JM, Serra-Mitjà P, Ferreiro L, Botana-Rial M, Ramos-Hernández C, Brenes JM, Canales L, Camacho V, Romero-Romero B, Trujillo JC, Martinez E, Cases E, Barba A, Majem M, Güell E, Pajares V. Malignant Pleural Effusion: A Multidisciplinary Approach. OPEN RESPIRATORY ARCHIVES 2024; 6:100349. [PMID: 39091982 PMCID: PMC11293617 DOI: 10.1016/j.opresp.2024.100349] [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: 04/22/2024] [Accepted: 06/10/2024] [Indexed: 08/04/2024] Open
Abstract
Malignant pleural effusion (MPE) has become an increasingly prevalent complication in oncological patients, negatively impacting their quality of life and casting a shadow over their prognosis. Owing to the pathophysiological mechanisms involved and the heterogeneous nature of the underlying disease, this entity is both a diagnostic and therapeutic challenge. Advances in the understanding of MPE have led to a shift in the treatment paradigm towards a more personalized approach. This article provides a comprehensive review and update on the pathophysiology of MPE and describes the diagnostic tools and the latest advances in the treatment of this complex clinical entity.
Collapse
Affiliation(s)
- Ana Pardessus Otero
- Interventional Pulmonology, Respiratory Medicine Department, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
| | - Albert Rafecas-Codern
- Interventional Pulmonology, Respiratory Medicine Department, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
- Chronic Respiratory Disease Group (GREC), Institut de Recerca Sant Pau (IR SANT PAU), Spain
| | - José M. Porcel
- Pleural Medicine Unit, Department of Internal Medicine, Arnau de Vilanova University Hospital, IRBLleida, University of Lleida, Lleida, Spain
| | - Pere Serra-Mitjà
- Interventional Pulmonology, Respiratory Medicine Department, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
| | - Lucía Ferreiro
- Pulmonology Department, University Clinical Hospital of Santiago, Interdisciplinary Research Group in Pulmonology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Maribel Botana-Rial
- Broncopleural Unit, Pulmonary Deparment, Hospital Álvaro Cunqueiro, EOXI Vigo, PneumoVigoI+i Research Group, Sanitary Research Institute Galicia Sur (IISGS), Vigo, Spain
- CIBER de Enfermedades Respiratorias, Spain
| | - Cristina Ramos-Hernández
- Pulmonary Deparment, Hospital Álvaro Cunqueiro, EOXI Vigo, PneumoVigoI+i Research Group, Sanitary Research Institute Galicia Sur (IISGS), Vigo, Spain
| | - José Manuel Brenes
- Radiology Department, Hospital Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
| | - Lydia Canales
- Radiology Department, Hospital Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
| | - Valle Camacho
- Nuclear Medicine Department, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Juan Carlos Trujillo
- Department of Thoracic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Elisabeth Martinez
- Department of Thoracic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Enrique Cases
- Interventional Pulmonology, Hospital Universitario Politécnico La Fe, Valencia, Spain
| | - Andrés Barba
- Medical Oncology Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Margarita Majem
- Medical Oncology Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Ernest Güell
- Palliative Care Unit, Oncology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma Barcelona (UAB), Barcelona, Spain
| | - Virginia Pajares
- Interventional Pulmonology, Respiratory Medicine Department, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma Barcelona (UAB), Barcelona, Spain
- Chronic Respiratory Disease Group (GREC), Institut de Recerca Sant Pau (IR SANT PAU), Spain
| |
Collapse
|
57
|
Ascierto PA, Mandalà M, Ferrucci PF, Guidoboni M, Rutkowski P, Ferraresi V, Arance A, Guida M, Maiello E, Gogas H, Richtig E, Quaglino P, Lebbé C, Helgadottir H, Queirolo P, Spagnolo F, Tucci M, Del Vecchio M, Gonzalez-Cao M, Minisini AM, De Placido S, Sanmamed MF, Casula M, Bulgarelli J, Pisano M, Piccinini C, Piccin L, Cossu A, Mallardo D, Paone M, Vitale MG, Melero I, Grimaldi AM, Giannarelli D, Palmieri G, Dummer R, Sileni VC. Sequencing of Checkpoint or BRAF/MEK Inhibitors on Brain Metastases in Melanoma. NEJM EVIDENCE 2024; 3:EVIDoa2400087. [PMID: 39315864 DOI: 10.1056/evidoa2400087] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
BACKGROUND The impact of the order of treatment with checkpoint inhibitors or BRAF/MEK inhibitors on the development of brain metastases in patients with metastatic unresectable BRAFV600-mutant melanoma is unknown. The SECOMBIT trial examined the impact of the order of receipt of these treatments in such patients. METHODS In this three-arm trial, we reviewed patients without brain metastases who received the BRAF/MEK inhibitors encorafenib and binimetinib until they had progressive disease followed by the immune checkpoint inhibitors ipilimumab and nivolumab (arm A); or treatment with ipilimumab and nivolumab until they had progressive disease followed by encorafenib and binimetinib (arm B); or treatment with encorafenib and binimetinib for 8 weeks followed by ipilimumab and nivolumab until they had progressive disease followed by retreatment with encorafenib arm binimetinib (arm C). RESULTS Brain metastases were discovered during the trial in 23/69 patients in arm A, 11/69 in arm B, and 9/68 in arm C. At a median follow-up of 56 months, the 60-month brain metastases-free survival rates were 56% for arm A, 80% for arm B (hazard ratio [HR] vs. A: 0.40, 95% confidence interval [CI] 0.23 to 0.58), and 85% for arm C (HR vs. A: 0.35, 95% CI 0.16 to 0.76). CONCLUSIONS In patients with unresectable metastatic melanoma, the treatment sequence of immune checkpoint inhibition followed by BRAF/MEK inhibitors was associated with longer periods of new brain metastases-free survival than the reverse sequence. A regimen in which immune checkpoint inhibition was sandwiched between BRAF/MEK inhibition also appeared to be protective against brain metastases. (ClinicalTrials.gov number NCT02631447.).
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Mario Mandalà
- Department of Oncologic, Hematologic, Gastroenterological Sciences, University of Perugia, Perugia, Italy
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | | | - Massimo Guidoboni
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska Curie National Research Institute of Oncology, 02-781, Warsaw, Poland
| | - Virginia Ferraresi
- Sarcomas and Rare Tumors Departmental Unit, IRCCS Regina Elena National Cancer Institute, Rome
| | - Ana Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona
| | - Michele Guida
- Rare Tumors and Melanoma Unit, IRCCS Istituto dei Tumori "Giovanni Paolo II," Bari, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens
| | - Erika Richtig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Céleste Lebbé
- Université Paris Cite, Dermato-Oncology and CIC AP-HP Hôpital Saint Louis, Cancer Institute APHP, Nord-Université Paris Cite, INSERM U976, Paris
| | - Hildur Helgadottir
- Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm
| | - Paola Queirolo
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Division of Melanoma, Sarcomas and Rare Tumors, IRCCS European Institute of Oncology, Milan
| | - Francesco Spagnolo
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genova, Italy
| | - Marco Tucci
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro," Bari, Italy
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Maria Gonzalez-Cao
- Department of Medical Oncology, Oncology Institute Rosell, University Hospital Dexeus, Barcelona
| | - Alessandro Marco Minisini
- Academic Hospital "Santa Maria della Misericordia," Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Miguel F Sanmamed
- Department of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Milena Casula
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Jenny Bulgarelli
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Marina Pisano
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Claudia Piccinini
- Experimental and Clinical Oncology of Immunotherapy and Rare Cancers Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori," Meldola, Italy
| | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Antonio Cossu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Domenico Mallardo
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Miriam Paone
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Maria Grazia Vitale
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
| | - Ignacio Melero
- Department of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Antonio M Grimaldi
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori - IRCCS Fondazione "G. Pascale," Napoli, Italy
- Medical Oncology Unit, AORN San Pio, Benevento, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome
| | - Giuseppe Palmieri
- Immuno-Oncology & Targeted Cancer Biotherapies, Unit of Cancer Genetics, University of Sassari, IRGB-CNR, 07100 Sassari, Italy
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
58
|
Lee Boniao E, Allen RC, Sundar G. Targeted therapy and immunotherapy for orbital and periorbital tumors: a major review. Orbit 2024; 43:656-673. [PMID: 37728602 DOI: 10.1080/01676830.2023.2256848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023]
Abstract
Traditionally, for patients who are poor candidates for surgery and/or radiotherapy, palliative chemotherapy is often offered but with significant toxic side effects. However, recent advancements in our understanding of tumor biology and molecular genetics have brought new understanding to the molecular pathways of certain tumors and cancers. This has ushered in a new era of precision medicine specific to a tumor or cancer treatment pathway (targeted therapy) or directed to host-tumor responses (immunotherapy). This article will focus on recent updates in the application of available targeted and immunotherapy for managing orbital and periorbital tumors and tumor-like conditions, which include cutaneous basal cell carcinoma, cutaneous squamous cell carcinoma, cutaneous melanoma, Merkel cell carcinoma, sebaceous gland carcinoma, solitary fibrous tumor, dermatofibrosarcoma protuberans, orbital meningioma, neurofibromatosis, Langerhans cell histiocytosis, ocular adnexal lymphoma, orbital lymphatic malformation, and adenoid cystic carcinoma.
Collapse
Affiliation(s)
- Emmanuel Lee Boniao
- Orbit & Oculofacial Surgery, Ophthalmic Oncology, Department of Ophthalmology, National University Hospital, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, Amai Pakpak Medical Center, Marawi City, Philippines
| | - Richard C Allen
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Gangadhara Sundar
- Orbit & Oculofacial Surgery, Ophthalmic Oncology, Department of Ophthalmology, National University Hospital, National University of Singapore, Singapore, Singapore
| |
Collapse
|
59
|
Taglialatela I, Indini A, Santanelli G, Di Liberti G, Di Guardo L, De Braud F, Del Vecchio M. Melanoma and sex hormones: Pathogenesis, progressive disease and response to treatments. TUMORI JOURNAL 2024; 110:309-318. [PMID: 38372040 DOI: 10.1177/03008916241231687] [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] [Indexed: 02/20/2024]
Abstract
Cutaneous melanoma represents the fifth tumor in terms of incidence in young adults, with a major involvement of males than females. Despite the significant changes in available effective treatments for cutaneous melanoma, there is still a proportion of patients that do not benefit long-term disease control with immune checkpoint inhibitors and/or BRAF/MEK inhibitors, and eventually develop progressive disease. In addition to the emerging biomarkers under investigation to understand resistance to treatments, recent studies resumed the role of sex hormones (estrogens, progesterone and androgens) in melanoma patients. In the last decades, the impact of sex hormones has been considered controversial in melanoma patients, but actual growing preclinical and clinical evidence underline the potential influence on melanoma cells' growth, tumor microenvironment, the immune system and consequently on the course of disease.This review will provide available insights on the role of sex hormones in melanoma pathogenesis, disease progression and response/resistance to systemic treatments. We will also offer an overview on the recent studies on the theme, describing the hormonal contribution to disease response and the interaction with targeted therapies and immune-checkpoint inhibitors in cutaneous melanoma patients, illustrating an insight into future research in this field.
Collapse
Affiliation(s)
- Ida Taglialatela
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alice Indini
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giulia Santanelli
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giorgia Di Liberti
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Lorenza Di Guardo
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Filippo De Braud
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
- Università degli studi di Milano, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michele Del Vecchio
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
60
|
Wang P, Yuan Y, Yang T, Zou Y, Tang M, Ma Z, Bo W, Qin S, Chen Y, Guo T, Guo Z, Yang J, Xiang M, Chen L. Design, Synthesis, and Biological Evaluation of 5-Amino-4-fluoro-1 H-benzo[ d]imidazole-6-carboxamide Derivatives as Novel and Potential MEK/RAF Complex Inhibitors Based on the "Clamp" Strategy. J Med Chem 2024; 67:15246-15267. [PMID: 39166848 DOI: 10.1021/acs.jmedchem.4c00860] [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: 08/23/2024]
Abstract
Herein, we described the rational drug design and synthesis of a series of 5-amino-4-fluoro-1H-benzo[d]imidazole-6-carboxamide derivatives that inhibit MEK and RAF kinases. The detailed screening cascades revealed that 16b was a preferred compound, which might act like a "clamp" to stabilize the MEK/RAF complex, thereby effectively inhibiting MEK1, BRAF, and BRAFV600E with IC50 values of 28, 3, and 3 nM, respectively. 16b possessed an excellent selectivity over other 312 human-related kinases at 1 μM. In vitro, 16b showed potent antiproliferative activities against MIA PaCa-2 (G12C KRAS), HCT116 (G13D KRAS), and C26 (G12D KRAS) cells with IC50 values of 0.011, 0.079, and 0.096 μM, respectively. CoIP experiments demonstrated that 16b could induce MEK/RAF complex formation. Most importantly, in the C26 syngeneic colorectal and HCT116 mice xenograft tumor models, 16b demonstrated tumor growth inhibition of 70 and 93%, respectively, suggesting that 16b may be a promising MEK/RAF complex inhibitor and worthy of further development.
Collapse
Affiliation(s)
- Peng Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongting Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yurong Zou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyan Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weichen Bo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Songhui Qin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongning Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianhong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingli Xiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
61
|
Chen MF, Yang SR, Tao JJ, Desilets A, Diamond EL, Wilhelm C, Rosen E, Gong Y, Mullaney K, Torrisi J, Young RJ, Somwar R, Yu HA, Kris MG, Riely GJ, Arcila ME, Ladanyi M, Donoghue MT, Rosen N, Yaeger R, Drilon A, Murciano-Goroff YR, Offin M. Tumor-Agnostic Genomic and Clinical Analysis of BRAF Fusions Identifies Actionable Targets. Clin Cancer Res 2024; 30:3812-3823. [PMID: 38922339 PMCID: PMC11371517 DOI: 10.1158/1078-0432.ccr-23-3981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/18/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Even though BRAF fusions are increasingly detected in standard multigene next-generation sequencing panels, few reports have explored their structure and impact on clinical course. EXPERIMENTAL DESIGN We collected data from patients with BRAF fusion-positive cancers identified through a genotyping protocol of 97,024 samples. Fusions were characterized and reviewed for oncogenic potential (in-frame status, non-BRAF partner gene, and intact BRAF kinase domain). RESULTS We found 241 BRAF fusion-positive tumors from 212 patients with 82 unique 5' fusion partners spanning 52 histologies. Thirty-nine fusion partners were not previously reported, and 61 were identified once. BRAF fusion incidence was enriched in pilocytic astrocytomas, gangliogliomas, low-grade neuroepithelial tumors, and acinar cell carcinoma of the pancreas. Twenty-four patients spanning multiple histologies were treated with MAPK-directed therapies, of which 20 were evaluable for RECIST. Best response was partial response (N = 2), stable disease (N = 11), and progressive disease (N = 7). The median time on therapy was 1 month with MEK plus BRAF inhibitors [(N = 11), range 0-18 months] and 8 months for MEK inhibitors [(N = 14), range 1-26 months]. Nine patients remained on treatment for longer than 6 months [pilocytic astrocytomas (N = 6), Erdheim-Chester disease (N = 1), extraventricular neurocytoma (N = 1), and melanoma (N = 1)]. Fifteen patients had acquired BRAF fusions. CONCLUSIONS BRAF fusions are found across histologies and represent an emerging actionable target. BRAF fusions have a diverse set of fusion partners. Durable responses to MAPK therapies were seen, particularly in pilocytic astrocytomas. Acquired BRAF fusions were identified after targeted therapy, underscoring the importance of postprogression biopsies to optimize treatment at relapse in these patients.
Collapse
Affiliation(s)
- Monica F. Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica J. Tao
- Department of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Antoine Desilets
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eli L. Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Clare Wilhelm
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ezra Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Yixiao Gong
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kerry Mullaney
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jean Torrisi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J. Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helena A. Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mark G. Kris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Gregory J. Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria E. Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark T.A. Donoghue
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Yonina R. Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
62
|
Nakano E, Takahashi A, Ogata D, Namikawa K, Yamazaki N. Real-world efficacy and safety of BRAF-targeted therapy for patients with advanced melanoma: A single-center retrospective study in Japan. J Dermatol 2024; 51:1199-1207. [PMID: 38716655 DOI: 10.1111/1346-8138.17263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/10/2024] [Accepted: 04/21/2024] [Indexed: 09/03/2024]
Abstract
Most clinical trials investigating targeted therapies for patients harboring BRAF V600 mutations have included mostly White patients, and data for Asian patients are scarce. Although there are several retrospective studies in Japanese patients, they have investigated only the dabrafenib + trametinib regimen, and have had a short follow-up period. We conducted a single-center retrospective study to update previous studies and compare the outcomes with those in White patients. We analyzed 89 patients who received dabrafenib + trametinib or encorafenib + binimetinib, including 11 who received both treatment regimens. The overall response rate was 79.8%, with complete response in 25 patients (28.1%) and partial response in 45 patients (51.7%). The median progression-free survival was 13.7 months, and the median overall survival was 32.9 months. The 3-year progression-free and overall survival rates were 31.8% and 47.9%, respectively. Although the two regimens showed no significant differences in efficacy, their safety profiles differed, as reported in clinical trials. Therefore, the most frequent adverse event associated with the dabrafenib + trametinib regimen was pyrexia (61.3%) and that of encorafenib + binimetinib was blurred vision (32.0%). Switching directly to another targeted therapy after progressive disease showed no clinical response; however, rechallenge followed by immune checkpoint inhibitor therapy showed a certain response. As a prognostic factor, performance status was associated with progression-free survival, and performance status, serum lactate dehydrogenase level, and dose interruption were associated with overall survival in the multivariate analysis. Real-world data on targeted therapy for patients with melanoma in Japan suggest that both dabrafenib + trametinib and encorafenib + binimetinib show similar efficacy and safety in Asian and White patients.
Collapse
Affiliation(s)
- Eiji Nakano
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Takahashi
- Department of Dermatologic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
63
|
Arghidash F, Javid-Naderi MJ, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Exploring the multifaceted effects of silymarin on melanoma: Focusing on the role of lipid-based nanocarriers. J Drug Deliv Sci Technol 2024; 99:105950. [DOI: 10.1016/j.jddst.2024.105950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
64
|
Gallois C, Bergen ES, Auclin É, Pernot S, Higué J, Trouilloud I, Touchefeu Y, Turpin A, Mazard T, Sartore-Bianchi A, Prenen H, Alberti A, Pilla L, Cuissy S, Wookey V, Perret A, Melchior C, Artru P, Dubreuil O, Drouillard A, Doat S, Lavolé J, Basile D, Perkins G, Jary M, Stintzing S, Ros J, Tougeron D, Taieb J. Efficacy and safety of the combination of encorafenib/cetuximab with or without binimetinib in patients with BRAF V600E-mutated metastatic colorectal cancer: an AGEO real-world multicenter study. ESMO Open 2024; 9:103696. [PMID: 39255538 PMCID: PMC11415680 DOI: 10.1016/j.esmoop.2024.103696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The combination of encorafenib with cetuximab has become the standard of care in patients with BRAF V600E-mutated metastatic colorectal cancer (mCRC) after a prior systemic therapy. This study aims to describe the efficacy and safety of encorafenib/cetuximab +/- binimetinib in patients with BRAF V600E-mutated mCRC in a real-world setting. PATIENTS AND METHODS This retrospective study included patients with BRAF V600E-mutated mCRC who received this combination from January 2020 to June 2022 in 30 centers. RESULTS A total of 201 patients were included, with 55% of women, a median age of 62 years, and an Eastern Cooperative Oncology Group performance status (ECOG-PS) >1 in 20% of cases. The main tumor characteristics were 60% of right-sided primary tumor, 11% of microsatellite instability/mismatch repair deficient phenotype, and liver and peritoneum being the two main metastatic sites (57% and 51%). Encorafenib/cetuximab +/- binimetinib was prescribed in the first, second, third, and beyond third line in 4%, 56%, 29%, and 11%, respectively, of cases, with the encorafenib/cetuximab/binimetinib combination for 21 patients (10%). With encorafenib/cetuximab treatment, 21% of patients experienced grade ≥3 adverse events (AEs), with each type of grade ≥3 AE observed in <5% of patients. The objective response rate was 32.2% and the disease control rate (DCR) was 71.2%. The median progression-free survival (PFS) was 4.5 months [95% confidence interval (CI) 3.9-5.4 months] and the median overall survival (OS) was 9.2 months (95% CI 7.8-10.8 months). In multivariable analysis, factors associated with a shorter PFS were synchronous metastases [hazard ratio (HR) 1.66, P = 0.04] and ECOG-PS >1 (HR 1.88, P = 0.007), and those associated with a shorter OS were the same factors (HR 1.71, P = 0.03 and HR 2.36, P < 0.001, respectively) in addition to treatment beyond the second line (HR 1.74, P = 0.003) and high carcinoembryonic antigen level (HR 1.72, P = 0.003). CONCLUSION This real-world study showed that in patients with BRAF V600E-mutated mCRC treated with encorafenib/cetuximab +/- binimetinib, efficacy and safety data confirm those reported in the BEACON registration trial. The main poor prognostic factors for this treatment are synchronous metastases and ECOG-PS >1.
Collapse
Affiliation(s)
- C Gallois
- Department of Gastroenterology and Digestive Oncology, Paris-Cité University, Georges Pompidou European Hospital, SIRIC CARPEM, Paris, France
| | - E S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - É Auclin
- Medical and Thoracic Oncology Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - S Pernot
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - J Higué
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - I Trouilloud
- Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Y Touchefeu
- Digestive Oncology, Institut Des Maladies De l'Appareil Digestif, Centre Hospitalier Universitaire De Nantes, Nantes, France
| | - A Turpin
- Department of Medical Oncology, University Lille, Lille, France; UMR9020 CNRS, UMR-S1277 Inserm, Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, CHU Lille, Lille, France
| | - T Mazard
- Department of Medical Oncology, Montpellier Cancer Institute (ICM), Montpellier, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Montpellier, France
| | - A Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano and Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - H Prenen
- University Hospital Antwerp, Edegem, Belgium
| | - A Alberti
- Medical Oncology, University of Brescia, ASST-Spedali Civili, Brescia, Italy
| | - L Pilla
- Department of Gastroenterology and Digestive Oncology, Paris-Cité University, Georges Pompidou European Hospital, SIRIC CARPEM, Paris, France
| | - S Cuissy
- Department of Hepatogastroenterology, Rouen University Hospital, Rouen, France
| | - V Wookey
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - A Perret
- Department of Medical Oncology, Gustave Roussy Cancer Centre, Villejuif, France
| | - C Melchior
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - P Artru
- Hepatogastroenterology Department, Hôpital Jean-Mermoz, Lyon, France
| | - O Dubreuil
- Department of Digestive Oncology, Groupe hospitalier Diaconesses Croix Saint Simon, Paris, France
| | - A Drouillard
- Department of Hepato-Gastroenterology, Dijon Hospital, Dijon, France
| | - S Doat
- Department of Hepato-Gastroenterology, Pitié-Salpêtrière Hospital, Paris, France
| | - J Lavolé
- Department of Hepato-Gastroenterology, Begin Teaching Military Hospital, Saint-Mandé, France
| | - D Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, Crotone, Italy
| | - G Perkins
- Department of Gastroenterology, CHRU Pontchaillou, Rennes, France
| | - M Jary
- Department of Surgical and Medical Oncology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - S Stintzing
- Department of Hematology, Oncology, and Cancer Immunology (CCM), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - J Ros
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - D Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - J Taieb
- Department of Gastroenterology and Digestive Oncology, Paris-Cité University, Georges Pompidou European Hospital, SIRIC CARPEM, Paris, France.
| |
Collapse
|
65
|
Nakamura K, Yaguchi T, Murata M, Ota Y, Mikoshiba A, Kiniwa Y, Okuyama R, Kawakami Y. Tumor eradication by triplet therapy with BRAF inhibitor, TLR 7 agonist, and PD-1 antibody for BRAF-mutated melanoma. Cancer Sci 2024; 115:2879-2892. [PMID: 38894534 PMCID: PMC11462939 DOI: 10.1111/cas.16251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Programmed death 1 (PD-1)/programmed death-ligand 1 inhibitors are commonly used to treat various cancers, including melanoma. However, their efficacy as monotherapy is limited, and combination immunotherapies are being explored to improve outcomes. In this study, we investigated a combination immunotherapy involving an anti-PD-1 antibody that blocks the major adaptive immune-resistant mechanisms, a BRAF inhibitor that inhibits melanoma cell proliferation, and multiple primary immune-resistant mechanisms, such as cancer cell-derived immunosuppressive cytokines, and a Toll-like receptor 7 agonist that enhances innate immune responses that promote antitumor T-cell induction and functions. Using a xenogeneic nude mouse model implanted with human BRAF-mutated melanoma, a BRAF inhibitor vemurafenib was found to restore T-cell-stimulatory activity in conventional dendritic cells by reducing immunosuppressive cytokines, including interleukin 6, produced by human melanoma. Additionally, intravenous administration of the Toll-like receptor 7 agonist DSR6434 enhanced tumor growth inhibition by vemurafenib through stimulating the plasmacytoid dendritic cells/interferon-α/natural killer cell pathways and augmenting the T-cell-stimulatory activity of conventional dendritic cells. In a syngeneic mouse model implanted with murine BRAF-mutated melanoma, the vemurafenib and DSR6434 combination synergistically augmented the induction of melanoma antigen gp100-specific T cells and inhibited tumor growth. Notably, only triplet therapy with vemurafenib, DSR6434, and the anti-PD-1 antibody resulted in complete regression of SIY antigen-transduced BRAF-mutated melanoma in a CD8 T-cell-dependent manner. These findings indicate that a triple-combination strategy targeting adaptive and primary resistant mechanisms while enhancing innate immune responses that promote tumor-specific T cells may be crucial for effective tumor eradication.
Collapse
Affiliation(s)
- Kenta Nakamura
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of MedicineKeio UniversityTokyoJapan
- Department of DermatologyShinshu University School of MedicineNaganoJapan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of MedicineKeio UniversityTokyoJapan
- Department of Immunology and Genomic MedicineKyoto University Graduate School of MedicineKyotoJapan
| | | | - Yosuke Ota
- Cancer Research UnitSumitomo Pharma Co. Ltd.OsakaJapan
| | - Asuka Mikoshiba
- Department of DermatologyShinshu University School of MedicineNaganoJapan
| | - Yukiko Kiniwa
- Department of DermatologyShinshu University School of MedicineNaganoJapan
| | - Ryuhei Okuyama
- Department of DermatologyShinshu University School of MedicineNaganoJapan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of MedicineKeio UniversityTokyoJapan
- Department of Immunology, School of MedicineInternational University of Health and WelfareChibaJapan
| |
Collapse
|
66
|
Wu W, Wu MY, Dai T, Ke LN, Shi Y, Hu J, Wang Q. Terphenyllin induces CASP3-dependent apoptosis and pyroptosis in A375 cells through upregulation of p53. Cell Commun Signal 2024; 22:409. [PMID: 39169379 PMCID: PMC11337594 DOI: 10.1186/s12964-024-01784-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Melanoma, one of the most lethal forms of skin cancer, has the potential to develop in any area where melanocytes are present. Currently, postoperative recurrence due to the emergence of systemic drug resistance represents a significant challenge in the treatment of melanoma. In this study, terphenyllin (TER), a distinctive inhibitory impact on melanoma cells was identified from the natural p-terphenyl metabolite. This study aimed to elucidate the intrinsic mechanism of this inhibitory effect, which may facilitate the discovery of novel chemotherapeutic agents. METHODS A transcriptome sequencing and metabolomic analysis of TER-treated A375 cells was conducted to identify potential pathways of action. The key proteins were knocked out and backfilled using CRISPR-Cas9 technology and molecular cloning. Subsequently, the results of cytosolic viability, LDH release, immunofluorescence and flow cytometry were employed to demonstrate the cell death status of the drug-treated cells. RESULTS The p53 signalling pathway was markedly upregulated following TER treatment, leading to the activation of CASP3 via the intrinsic apoptotic pathway. The activated CASP3 initiated apoptosis, while simultaneously continuing to cleave the GSDME, thereby triggering pyroptosis. The knockout of p53, a key protein situated upstream of this pathway, resulted in a significant rescue of TER-induced cell death, as well as an alleviation of the decrease in cell viability. However, the knockout of key proteins situated downstream of the pathway (CASP3 and GSDME) did not result in a rescue of TER-induced cell death, but rather a transformation of the cells from apoptosis and pyroptosis. CONCLUSIONS The induction of apoptosis and pyroptosis in A375 cells by TER is mediated via the p53-BAX/FAS-CASP3-GSDME signalling pathway. This lays the foundation for TER as a potential anti-melanoma drug in the future. It should be noted that CASP3 and GSDME in this pathway solely regulate the mode of cell death, rather than determine whether cell death occurs. This distinction may prove valuable in future studies of apoptosis and pyroptosis.
Collapse
Affiliation(s)
- Wei Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Meng-Yuan Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Ting Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Li-Na Ke
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Yan Shi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jin Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
67
|
Mechahougui H, Gutmans J, Colarusso G, Gouasmi R, Friedlaender A. Advances in Personalized Oncology. Cancers (Basel) 2024; 16:2862. [PMID: 39199633 PMCID: PMC11352922 DOI: 10.3390/cancers16162862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Advances in next-generation sequencing (NGS) have catalyzed a paradigm shift in cancer treatment, steering the focus from conventional, organ-specific protocols to precision medicine. Emerging targeted therapies offer a cutting-edge approach to cancer treatment, while companion diagnostics play an essential role in aligning therapeutic choices with specific molecular changes identified through NGS. Despite these advances, interpreting the clinical implications of a rapidly expanding catalog of genetic mutations remains a challenge. The selection of therapies in the presence of multiple mutations requires careful clinical judgment, supported by quality-centric genomic testing that emphasizes actionable mutations. Molecular tumor boards can play an increasing role in assimilating genomic data into clinical trials, thereby refining personalized treatment approaches and improving patient outcomes.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - Gina Colarusso
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (H.M.)
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France
| | | |
Collapse
|
68
|
Dastgheib ZS, Abolmaali SS, Farahavar G, Salmanpour M, Tamaddon AM. Gold nanostructures in melanoma: Advances in treatment, diagnosis, and theranostic applications. Heliyon 2024; 10:e35655. [PMID: 39170173 PMCID: PMC11336847 DOI: 10.1016/j.heliyon.2024.e35655] [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: 02/15/2024] [Revised: 06/16/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Melanoma, a lethal form of skin cancer, poses a significant challenge in oncology due to its aggressive nature and high mortality rates. Gold nanostructures, including gold nanoparticles (GNPs), offer myriad opportunities in melanoma therapy and imaging due to their facile synthesis and functionalization, robust stability, tunable physicochemical and optical properties, and biocompatibility. This review explores the emerging role of gold nanostructures and their composites in revolutionizing melanoma treatment paradigms, bridging the gap between nanotechnology and clinical oncology, and offering insights for researchers, clinicians, and stakeholders. It begins by elucidating the potential of nanotechnology-driven approaches in cancer therapy, highlighting the unique physicochemical properties and versatility of GNPs in biomedical applications. Various therapeutic modalities, including photothermal therapy, photodynamic therapy, targeted drug delivery, gene delivery, and nanovaccines, are discussed in detail, along with insights from ongoing clinical trials. In addition, the utility of GNPs in melanoma imaging and theranostics is explored, showcasing their potential in diagnosis, treatment monitoring, and personalized medicine. Furthermore, safety considerations and potential toxicities associated with GNPs are addressed, underscoring the importance of comprehensive risk assessment in clinical translation. Finally, the review concludes by discussing current challenges and future directions, emphasizing the need for innovative strategies to maximize the clinical impact of GNPs in melanoma therapy.
Collapse
Affiliation(s)
- Zahra Sadat Dastgheib
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| | - Mohsen Salmanpour
- Cellular and Molecular Biology Research Center, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Ali Mohammad Tamaddon
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, 71345, Iran
| |
Collapse
|
69
|
van Not OJ, van den Eertwegh AJM, Jalving H, Bloem M, Haanen JB, van Rijn RS, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, de Groot J. W. B. JW, Hospers GAP, Kapiteijn E, Leeneman B, D. P, Stevense-den Boer M, van der Veldt AAM, Vreugdenhil G. G, Wouters MWJM, Blokx WAM, Suijkerbuijk KPM. Long-Term Survival in Patients With Advanced Melanoma. JAMA Netw Open 2024; 7:e2426641. [PMID: 39141388 PMCID: PMC11325208 DOI: 10.1001/jamanetworkopen.2024.26641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/31/2024] [Indexed: 08/15/2024] Open
Abstract
IMPORTANCE Long-term survival data from clinical trials show that survival curves of patients with advanced melanoma treated with immune checkpoint inhibitors (ICIs) gradually reach a plateau, suggesting that patients have a chance of achieving long-term survival. OBJECTIVE To investigate long-term survival in patients with advanced melanoma treated with ICIs outside clinical trials. DESIGN, SETTING, AND PARTICIPANTS Cohort study using prospectively collected data from the nationwide Dutch Melanoma Treatment Registry, including patients in the Netherlands with advanced melanoma treated with first-line ICIs from 2012 to 2019. Data were analyzed from January to September 2023. EXPOSURES Patients were treated with first-line ipilimumab-nivolumab, antibodies that target programmed cell death (anti-PD-1), or ipilimumab. MAIN OUTCOMES AND MEASURES Progression-free survival (PFS) and melanoma-specific survival were analyzed, and a Cox proportional hazards model was used to investigate factors associated with PFS after reaching partial response (PR) or complete response (CR). RESULTS A total of 2490 patients treated with first-line ICIs were included (median [IQR] age, 65.0 [55.3-73.0] years; 1561 male patients [62.7%]). Most patients had an Eastern Cooperative Oncology Group Performance Status of 1 or lower (2202 patients [88.5%]) and normal lactate dehydrogenase levels (1715 patients [68.9%]). PFS for all patients was 23.4% (95% CI, 21.7%-25.2%) after 3 years and 19.7% (95% CI, 18.0%-21.4%) after 5 years. Overall survival for all patients was 44.0% (95% CI, 42.1%-46.1%) after 3 years and 35.9% (95% CI, 33.9%-38.0%) after 5 years. Patients with metastases in 3 or more organ sites had a significantly higher hazard of progression after reaching PR or CR (adjusted hazard ratio, 1.37; 95% CI, 1.11-1.69). CONCLUSIONS AND RELEVANCE This cohort study of patients with advanced melanoma treated with ICIs in clinical practice showed that their survival reached a plateau, comparable with patients participating in clinical trials. These findings can be used in daily clinical practice to guide long-term surveillance strategies and inform both physicians and patients regarding long-term treatment outcomes.
Collapse
Affiliation(s)
- Olivier J. van Not
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Alfons J. M. van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Manja Bloem
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B. Haanen
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Maureen J. B. Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | | | - Christian U. Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marye J. Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geke A. P. Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Brenda Leeneman
- Department of Health Technology Assessment, Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
- Erasmus Center for Health Economics Rotterdam, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Piersma D.
- Department of Internal Medicine, Medisch Spectrum Twente, Enschede, the Netherlands
| | | | - Astrid A. M. van der Veldt
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Michel W. J. M. Wouters
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, the Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willeke A. M. Blokx
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karijn P. M. Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
70
|
Bromberger S, Zadorozhna Y, Ressler JM, Holzner S, Nawrocki A, Zila N, Springer A, Røssel Larsen M, Schossleitner K. Off-targets of BRAF inhibitors disrupt endothelial signaling and vascular barrier function. Life Sci Alliance 2024; 7:e202402671. [PMID: 38839106 PMCID: PMC11153892 DOI: 10.26508/lsa.202402671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
Targeted therapies against mutant BRAF are effectively used in combination with MEK inhibitors (MEKi) to treat advanced melanoma. However, treatment success is affected by resistance and adverse events (AEs). Approved BRAF inhibitors (BRAFi) show high levels of target promiscuity, which can contribute to these effects. The blood vessel lining is in direct contact with high plasma concentrations of BRAFi, but effects of the inhibitors in this cell type are unknown. Hence, we aimed to characterize responses to approved BRAFi for melanoma in the vascular endothelium. We showed that clinically approved BRAFi induced a paradoxical activation of endothelial MAPK signaling. Moreover, phosphoproteomics revealed distinct sets of off-targets per inhibitor. Endothelial barrier function and junction integrity were impaired upon treatment with vemurafenib and the next-generation dimerization inhibitor PLX8394, but not with dabrafenib or encorafenib. Together, these findings provide insights into the surprisingly distinct side effects of BRAFi on endothelial signaling and functionality. Better understanding of off-target effects could help to identify molecular mechanisms behind AEs and guide the continued development of therapies for BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Sophie Bromberger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Yuliia Zadorozhna
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Silvio Holzner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Arkadiusz Nawrocki
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- University of Applied Sciences FH Campus Wien, Division of Biomedical Science, Vienna, Austria
| | - Alexander Springer
- Department of Pediatric Surgery, Medical University of Vienna, Vienna, Austria
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
71
|
Kennedy LB, Salama AKS. Multiple Options: How to Choose Therapy in Frontline Metastatic Melanoma. Curr Oncol Rep 2024; 26:915-923. [PMID: 38837107 DOI: 10.1007/s11912-024-01547-0] [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] [Accepted: 05/10/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE OF REVIEW Given the rapid development of multiple targeted and immune therapies for patients with advanced melanoma, it can be challenging to select a therapy based on currently available data. This review aims to provide an overview of frontline options for metastatic melanoma, with practical guidance for selecting a treatment regimen. RECENT FINDINGS Recently reported data from randomized trials suggests that the majority of patients with unresectable melanoma should receive a PD-1 checkpoint inhibitor as part of their first line therapy, irrespective of BRAF mutation status. Additional data also suggests that combination immunotherapies result in improved outcomes compared to single agent, albeit at the cost of increased toxicity, though to date no biomarker exists to help guide treatment selection. As the number therapeutic options continue to grow for patients with advanced melanoma, there is likely to be a continued focus on combination strategies. Defining the optimal treatment approach in order to maximize efficacy while minimizing toxicity remains an area of active investigation.
Collapse
Affiliation(s)
- Lucy Boyce Kennedy
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, 44195, USA
| | - April K S Salama
- Division of Medical Oncology, Duke University Hospital, Durham, NC, 27710, USA.
| |
Collapse
|
72
|
Embaby A, Huijberts SCFA, Wang L, Leite de Oliveira R, Rosing H, Nuijen B, Sanders J, Hofland I, van Steenis C, Kluin RJC, Lieftink C, Smith CG, Blank CU, van Thienen JV, Haanen JBAG, Steeghs N, Opdam FL, Beijnen JH, Huitema ADR, Bernards R, Schellens JHM, Wilgenhof S. A Proof-of-Concept Study of Sequential Treatment with the HDAC Inhibitor Vorinostat following BRAF and MEK Inhibitors in BRAFV600-Mutated Melanoma. Clin Cancer Res 2024; 30:3157-3166. [PMID: 38739109 DOI: 10.1158/1078-0432.ccr-23-3171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE The development of resistance limits the clinical benefit of BRAF and MEK inhibitors (BRAFi/MEKi) in BRAFV600-mutated melanoma. It has been shown that short-term treatment (14 days) with vorinostat was able to initiate apoptosis of resistant tumor cells. We aimed to assess the antitumor activity of sequential treatment with vorinostat following BRAFi/MEKi in patients with BRAFV600-mutated melanoma who progressed after initial response to BRAFi/MEKi. PATIENTS AND METHODS Patients with BRAFi/MEKi-resistant BRAFV600-mutated melanoma were treated with vorinostat 360 mg once daily for 14 days followed by BRAFi/MEKi. The primary endpoint was an objective response rate of progressive lesions of at least 30% according to Response Evaluation Criteria in Solid Tumors 1.1. Secondary endpoints included progression-free survival, overall survival, safety, pharmacokinetics of vorinostat, and translational molecular analyses using ctDNA and tumor biopsies. RESULTS Of the 26 patients with progressive BRAFi/MEKi-resistant BRAFV600-mutated melanoma receiving treatment with vorinostat, 22 patients were evaluable for response. The objective response rate was 9%, with one complete response for 31.2 months and one partial response for 14.9 months. Median progression-free survival and overall survival were 1.4 and 5.4 months, respectively. Common adverse events were fatigue (23%) and nausea (19%). ctDNA analysis showed emerging secondary mutations in NRAS and MEK in eight patients at the time of BRAFi/MEKi resistance. Elimination of these mutations by vorinostat treatment was observed in three patients. CONCLUSIONS Intermittent treatment with vorinostat in patients with BRAFi/MEKi-resistant BRAFV600-mutated melanoma is well tolerated. Although the primary endpoint of this study was not met, durable antitumor responses were observed in a minority of patients (9%).
Collapse
Affiliation(s)
- Alaa Embaby
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sanne C F A Huijberts
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rodrigo Leite de Oliveira
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- CEMM, Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Charlaine van Steenis
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Roelof J C Kluin
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Christian U Blank
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frans L Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos H Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | | | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
73
|
Kim KB. Personalized therapy in oncology: melanoma as a paradigm for molecular-targeted treatment approaches. Clin Exp Metastasis 2024; 41:465-471. [PMID: 38935186 DOI: 10.1007/s10585-024-10291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/28/2024]
Abstract
In recent decades, the field of systemic cancer treatment has seen remarkable changes due to advancements in the understanding of cancer's biology, immunology, and genetic makeup. As a result, individuals with late-stage cancers are now achieving survival rates that were previously unattainable. The goal of personalized cancer therapy is to enhance clinical outcomes by customizing drug treatments to suit the unique genetic and/or epigenetic profiles of each patient's tumor. This approach aims to reduce the side effects commonly associated with ineffective treatments. Advances in genetic sequencing and molecular cytogenetics have been instrumental in identifying cancer-driving mutations and epigenetic irregularities, leading to the development of specific molecular therapies. This review article highlights the progress and success of targeted molecular therapies in treating malignant melanoma, illustrating the concept of personalized cancer treatment.
Collapse
Affiliation(s)
- Kevin B Kim
- California Pacific Medical Center Research Institute, 2333 Buchanan St, San Francisco, CA, 94115, USA.
| |
Collapse
|
74
|
Wang M, Sullivan RJ, Mooradian MJ. Toxicities from BRAF and MEK Inhibitors: Strategies to Maximize Therapeutic Success. Curr Oncol Rep 2024; 26:934-944. [PMID: 38850505 DOI: 10.1007/s11912-024-01544-3] [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] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE OF REVIEW This report highlights several of the recent therapeutic advancements in the treatment of BRAF-mutant tumors, discusses the most common adverse events observed with BRAF-targeted agents, and suggests strategies to manage and mitigate treatment-related toxicities. RECENT FINDINGS BRAF and MEK inhibitors represent a significant advancement in the treatment of BRAF-mutated malignancies with data across tumor types demonstrating the anti-tumor efficacy of dual MAPK inhibition. Although these agents have a reasonable toxicity profile, variable side effects across organ systems can develop. The discovery of activating BRAF mutations and subsequent development of BRAF and MEK inhibitors has transformed the treatment algorithms of BRAF-mutant malignancies. With increased application of these targeted regimens, identification and prompt management of their unique adverse events are crucial.
Collapse
Affiliation(s)
- Mike Wang
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Meghan J Mooradian
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
75
|
Egeler MD, van Leeuwen M, Lai-Kwon J, Eriksson H, Bartula I, Elashwah S, Fox L, Van Hemelrijck M, Jefford M, Lijnsvelt J, Bagge ASL, Morag O, Ny L, Olofsson Bagge R, Rogiers A, Saw RPM, Serpentini S, Iannopollo L, Thompson J, Stiller HT, Vanlaer N, van Akkooi ACJ, van de Poll-Franse LV. Understanding quality of life issues in patients with advanced melanoma: Phase 1 and 2 in the development of the EORTC advanced melanoma module. Eur J Cancer 2024; 207:114176. [PMID: 38875843 DOI: 10.1016/j.ejca.2024.114176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024]
Abstract
AIMS We aimed to develop a European Organization for Research and Treatment of Cancer (EORTC) Quality of Life (QoL) module tailored for patients with advanced (resectable or unresectable stage III/IV) melanoma receiving immune checkpoint inhibitors or targeted therapy. METHODS Following the EORTC QoL Group module development guidelines, we conducted phases 1 and 2 of the development process. In phase 1, we generated a list of health-related (HR)QoL issues through a systematic literature review and semi-structured interviews with healthcare professionals (HCPs) and patients with advanced melanoma. In phase 2, these issues were converted into questionnaire items to create the preliminary module. RESULTS Phase 1: we retrieved 8006 articles for the literature review, of which 35 were deemed relevant, resulting in 84 HRQoL issues being extracted to create the initial issue list. Semi-structured interviews with 18 HCPs and 28 patients with advanced melanoma resulted in 28 issues being added to the initial issue list. Following EORTC module development criteria, 26 issues were removed, and two issues were added after review by patient advocates. Phase 2: To ensure uniformity and avoid duplication, 16 issues were consolidated into eight items. Additionally, an independent expert contributed one new item, resulting in a preliminary module comprising 80 HRQoL items. CONCLUSION We identified a range of HRQoL issues (dry skin, xerostomia, and arthralgia) relevant to patients with stage III/IV melanoma. Future module development phases will refine the questionnaire. Once completed, this module will enable standardized assessment of HRQoL in patients with (locally) advanced melanoma.
Collapse
Affiliation(s)
- M D Egeler
- Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - M van Leeuwen
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J Lai-Kwon
- Department of Medical Oncology and Department of Health Services Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - H Eriksson
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Sweden
| | - I Bartula
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - S Elashwah
- Medical Oncology Unit, Oncology Center, Mansoura University (OCMU), Egypt
| | - L Fox
- King's College London, London, United Kingdom
| | | | - M Jefford
- Department of Medical Oncology and Department of Health Services Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - J Lijnsvelt
- Netherlands Cancer Institute, Department of Medical Oncology, Amsterdam, the Netherlands
| | - A-S Lindqvist Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Psychology, University of Gothenburg, Gothenburg, Sweden
| | - O Morag
- Sheba Medical Center, The Jusjdman Cancer Center, Ramat-gan, Israel
| | - L Ny
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - R Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Rogiers
- Department of Medical Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - R P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - J Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | | | - N Vanlaer
- Sheba Medical Center, The Jusjdman Cancer Center, Ramat-gan, Israel
| | - A C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | |
Collapse
|
76
|
Paolino G, Pampena R, Di Ciaccio SM, Carugno A, Cantisani C, Di Nicola MR, Losco L, Bortone G, Mercuri SR, Costanzo A, Ardigò M, Valenti M. Thin Amelanotic and Hypomelanotic Melanoma: Clinicopathological and Dermoscopic Features. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1239. [PMID: 39202520 PMCID: PMC11356094 DOI: 10.3390/medicina60081239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Amelanotic/hypomelanotic melanomas (AHMs) account for 2-8% of all cutaneous melanomas. Due to their clinical appearance and the lack of specific dermoscopic indicators, AHMs are challenging to diagnose, particularly in thinner cutaneous lesions. The aim of our study was to evaluate the clinicopathological and dermoscopic features of thin AHMs. Identifying the baseline clinical-pathological features and dermoscopic aspects of thin AHMs is crucial to better understand this entity. Materials and Methods: We divided the AHM cohort into two groups based on Breslow thickness: thin (≤1.00 mm) and thick (>1.00 mm). This stratification helped identify any significant clinicopathological differences between the groups. For dermoscopic analysis, we employed the "pattern analysis" approach, which involves a simultaneous and subjective assessment of different criteria. Results: Out of the 2.800 melanomas analyzed for Breslow thickness, 153 were identified as AHMs. Among these, 65 patients presented with thin AHMs and 88 with thick AHMs. Red hair color and phototype II were more prevalent in patients with thin AHMs. The trunk was the most common anatomic site for thin AHMs. Patients with thin AHMs showed a higher number of multiple melanomas. Dermoscopic analysis revealed no significant difference between thin AHMs and thick AHMs, except for a more frequent occurrence of residual reticulum in thin AHMs. Conclusions: Thin AHMs typically affect individuals with lower phototypes and red hair color. These aspects can be related to the higher presence of pheomelanin, which provides limited protection against sun damage. This also correlates with the fact that the trunk, a site commonly exposed to intermittent sun exposure, is the primary anatomical location for thin AHMs. Multiple primary melanomas are more common in patients with thin AHMs, likely due to an intrinsic predisposition as well as greater periodic dermatologic follow-ups in this class of patients. Apart from the presence of residual reticulum, no other significant dermoscopic differences were observed, complicating the differential diagnosis between thin and thick AHMs based on dermoscopy alone.
Collapse
Affiliation(s)
- Giovanni Paolino
- Unit of Dermatology and Cosmetology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.P.); (S.R.M.)
| | - Riccardo Pampena
- La Sapienza University of Rome, 00185 Rome, Italy; (R.P.); (S.M.D.C.)
| | | | - Andrea Carugno
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Carmen Cantisani
- Dermatologic Clinic, La Sapienza University of Rome, 00185 Rome, Italy; (C.C.); (G.B.)
| | - Matteo Riccardo Di Nicola
- Unit of Dermatology and Cosmetology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.P.); (S.R.M.)
| | - Luigi Losco
- Plastic Surgery Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Baronissi, Italy;
| | - Giulio Bortone
- Dermatologic Clinic, La Sapienza University of Rome, 00185 Rome, Italy; (C.C.); (G.B.)
| | - Santo Raffaele Mercuri
- Unit of Dermatology and Cosmetology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.P.); (S.R.M.)
- UniSr Vita-Salute San Raffaele University, 20132 Milano, Italy
| | - Antonio Costanzo
- Dermatology Unit, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.C.); (M.A.); (M.V.)
| | - Marco Ardigò
- Dermatology Unit, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.C.); (M.A.); (M.V.)
| | - Mario Valenti
- Dermatology Unit, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.C.); (M.A.); (M.V.)
| |
Collapse
|
77
|
Boileve A, Smolenschi C, Lambert A, Boige V, Tarabay A, Valery M, Fuerea A, Pudlarz T, Conroy T, Hollebecque A, Ducreux M. Role of molecular biology in the management of pancreatic cancer. World J Gastrointest Oncol 2024; 16:2902-2914. [PMID: 39072173 PMCID: PMC11271790 DOI: 10.4251/wjgo.v16.i7.2902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 07/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges in patient management due to a dismal prognosis, increasing incidence, and limited treatment options. In this regard, precision medicine, which personalizes treatments based on tumour molecular characteristics, has gained great interest. However, its widespread implementation is not fully endorsed in current recommendations. This review explores key molecular alterations in PDAC, while emphasizing differences between KRAS-mutated and KRAS-wild-type tumours. It assesses the practical application of precision medicine in clinical settings and outlines potential future directions with respect to PDAC. Actionable molecular targets are examined with the aim of enhancing our understanding of PDAC molecular biology. Insights from this analysis may contribute to a more refined and personalized approach to pancreatic cancer treatment, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alice Boileve
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | | | - Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | - Valérie Boige
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Anthony Tarabay
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Marine Valery
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Alina Fuerea
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thomas Pudlarz
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thierry Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | | | - Michel Ducreux
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| |
Collapse
|
78
|
Deiana C, Agostini M, Brandi G, Giovannetti E. The trend toward more target therapy in pancreatic ductal adenocarcinoma. Expert Rev Anticancer Ther 2024; 24:525-565. [PMID: 38768098 DOI: 10.1080/14737140.2024.2357802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Despite the considerable progress made in cancer treatment through the development of target therapies, pancreatic ductal adenocarcinoma (PDAC) continues to exhibit resistance to this category of drugs. As a result, chemotherapy combination regimens remain the primary treatment approach for this aggressive cancer. AREAS COVERED In this review, we provide an in-depth analysis of past and ongoing trials on both well-known and novel targets that are being explored in PDAC, including PARP, EGFR, HER2, KRAS, and its downstream and upstream pathways (such as RAF/MEK/ERK and PI3K/AKT/mTOR), JAK/STAT pathway, angiogenesis, metabolisms, epigenetic targets, claudin, and novel targets (such as P53 and plectin). We also provide a comprehensive overview of the significant trials for each target, allowing a thorough glimpse into the past and future of target therapy. EXPERT OPINION The path toward implementing a target therapy capable of improving the overall survival of PDAC is still long, and it is unlikely that a monotherapy target drug will fulfill a meaningful role in addressing the complexity of this cancer. Thus, we discuss the future direction of target therapies in PDAC, trying to identify the more promising target and combination treatments, with a special focus on the more eagerly awaited ongoing trials.
Collapse
Affiliation(s)
- Chiara Deiana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Margherita Agostini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Associazione Italiana per la Ricerca sul Cancro (AIRC) Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, San Giuliano, Italy
| |
Collapse
|
79
|
Boutalaka M, El Bahi S, Alaqarbeh M, El Alaouy MA, Koubi Y, Khatabi KE, Maghat H, Bouachrine M, Lakhlifi T. Computational investigation of imidazo[2,1-b]oxazole derivatives as potential mutant BRAF kinase inhibitors: 3D-QSAR, molecular docking, molecular dynamics simulation, and ADMETox studies. J Biomol Struct Dyn 2024; 42:5268-5287. [PMID: 37424193 DOI: 10.1080/07391102.2023.2233629] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
BRAF inhibitors are known to be an effective therapeutic target for treating melanoma and other types of cancer. Using 3D-QSAR, molecular docking, and MD simulations, this study evaluated various imidazo[2,1-b]oxazole derivatives that function as mutant BRAF kinase inhibitors. Comparative molecular field analysis (CoMFA) and comparative molecular similarity index analysis (CoMSIA) were used to create the 3D-QSAR models. CoMSIA/SEHA model has solid predictive power across several models (Q2 = 0.578; R2 = 0.828; R2pred = 0.74) and is the best model according to the numerous field models generated. The created model's predictive power was evaluated through external validation using a test set. CoMSIA/SEHA contour maps collect information that can be used to identify critical regions with solid anticancer activity. We developed four inhibitors with high predicted activity due to these observations. ADMET prediction was used to assess the toxicity of the proposed imidazo[2,1-b]oxazole compounds. The predictive molecules (T1-T4) demonstrated good ADMET properties, excluding the toxic active compounds 11r from the database. Molecular docking was also used to determine the patterns and modes of interactions between imidazo[2,1-b]oxazole ligands and receptors, which revealed that the proposed imidazo[2,1-b]oxazole scaffold was stable in the receptor's active site (PDB code: 4G9C). The suggested compounds (T1-T4) were subjected to molecular dynamics simulations lasting 100 ns to determine their binding free energies. The results showed that T2 had a more favorable binding free energy (-149.552 kJ/mol) than T1 (-112.556 kJ/mol), T3 (-115.503 kJ/mol), and T4 (-102.553 kJ/mol). The results suggest that the imidazo[2,1-b]oxazole compounds investigated in this study have potential as inhibitors of BRAF kinase and could be further developed as anticancer drugs. Highlights22 imidazo[2,1-b]oxazole compounds were subjected to research on three-dimensional quantitative conformational relationships.Using contour maps from 3D-QSAR models as a guide was used to figure out the areas and strategies for structural optimization.Combined molecular docking, molecular dynamics simulations, and binding free energy calculations to verify the inhibitor activity of the proposed 22 imidazo[2,1-b]oxazole compounds.Four potential B-RAF Kinase inhibitors were discovered, providing theoretical clues for developing a highly anticancer agent.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Meryem Boutalaka
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Salma El Bahi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | | | - Moulay Ahfid El Alaouy
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Yassine Koubi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Khalil El Khatabi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Hamid Maghat
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| | - Mohammed Bouachrine
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
- EST Khenifra, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Tahar Lakhlifi
- Department of Chemistry, Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University of Moulay Ismail, Meknes, Morocco
| |
Collapse
|
80
|
Gu R, Fang H, Wang R, Dai W, Cai G. A comprehensive overview of the molecular features and therapeutic targets in BRAF V600E-mutant colorectal cancer. Clin Transl Med 2024; 14:e1764. [PMID: 39073010 PMCID: PMC11283586 DOI: 10.1002/ctm2.1764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
As one of the most prevalent digestive system tumours, colorectal cancer (CRC) poses a significant threat to global human health. With the emergence of immunotherapy and target therapy, the prognosis for the majority of CRC patients has notably improved. However, the subset of patients with BRAF exon 15 p.V600E mutation (BRAFV600E) has not experienced remarkable benefits from these therapeutic advancements. Hence, researchers have undertaken foundational investigations into the molecular pathology of this specific subtype and clinical effectiveness of diverse therapeutic drug combinations. This review comprehensively summarised the distinctive molecular features and recent clinical research advancements in BRAF-mutant CRC. To explore potential therapeutic targets, this article conducted a systematic review of ongoing clinical trials involving patients with BRAFV600E-mutant CRC.
Collapse
Affiliation(s)
- Ruiqi Gu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hongsheng Fang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Renjie Wang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Weixing Dai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Guoxiang Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
81
|
Karras F, Kunz M. Patient-derived melanoma models. Pathol Res Pract 2024; 259:155231. [PMID: 38508996 DOI: 10.1016/j.prp.2024.155231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024]
Abstract
Melanoma is a very aggressive, rapidly metastasizing tumor that has been studied intensively in the past regarding the underlying genetic and molecular mechanisms. More recently developed treatment modalities have improved response rates and overall survival of patients. However, the majority of patients suffer from secondary treatment resistance, which requires in depth analyses of the underlying mechanisms. Here, melanoma models based on patients-derived material may play an important role. Consequently, a plethora of different experimental techniques have been developed in the past years. Among these are 3D and 4D culture techniques, organotypic skin reconstructs, melanoma-on-chip models and patient-derived xenografts, Every technique has its own strengths but also weaknesses regarding throughput, reproducibility, and reflection of the human situation. Here, we provide a comprehensive overview of currently used techniques and discuss their use in different experimental settings.
Collapse
Affiliation(s)
- Franziska Karras
- Institute of Pathology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg 39120, Germany.
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University Medical Center Leipzig, Philipp-Rosenthal-Str. 23, Leipzig 04103, Germany
| |
Collapse
|
82
|
Kim M, Shim HS, Kim S, Lee IH, Kim J, Yoon S, Kim HD, Park I, Jeong JH, Yoo C, Cheon J, Kim IH, Lee J, Hong SH, Park S, Jung HA, Kim JW, Kim HJ, Cha Y, Lim SM, Kim HS, Lee CK, Kim JH, Chun SH, Yun J, Park SY, Lee HS, Cho YM, Nam SJ, Na K, Yoon SO, Lee A, Jang KT, Yun H, Lee S, Kim JH, Kim WS. Clinical practice recommendations for the use of next-generation sequencing in patients with solid cancer: a joint report from KSMO and KSP. J Pathol Transl Med 2024; 58:147-164. [PMID: 39026440 PMCID: PMC11261170 DOI: 10.4132/jptm.2023.11.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 07/20/2024] Open
Abstract
In recent years, next-generation sequencing (NGS)-based genetic testing has become crucial in cancer care. While its primary objective is to identify actionable genetic alterations to guide treatment decisions, its scope has broadened to encompass aiding in pathological diagnosis and exploring resistance mechanisms. With the ongoing expansion in NGS application and reliance, a compelling necessity arises for expert consensus on its application in solid cancers. To address this demand, the forthcoming recommendations not only provide pragmatic guidance for the clinical use of NGS but also systematically classify actionable genes based on specific cancer types. Additionally, these recommendations will incorporate expert perspectives on crucial biomarkers, ensuring informed decisions regarding circulating tumor DNA panel testing.
Collapse
Affiliation(s)
- Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - In Hee Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaekyung Cheon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sook Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Han Jo Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Yongjun Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jina Yun
- Division of Hematology/Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
83
|
Baik C, Cheng ML, Dietrich M, Gray JE, Karim NA. A Practical Review of Encorafenib and Binimetinib Therapy Management in Patients with BRAF V600E-Mutant Metastatic Non-Small Cell Lung Cancer. Adv Ther 2024; 41:2586-2605. [PMID: 38698170 PMCID: PMC11213720 DOI: 10.1007/s12325-024-02839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/08/2024] [Indexed: 05/05/2024]
Abstract
According to current guidelines, targeted therapy with a combination of BRAF plus MEK inhibitors is the preferred first-line treatment for patients with BRAF V600E-mutant metastatic non-small cell lung cancer (NSCLC). In the open-label, single-arm, phase 2 PHAROS trial (NCT03915951), the combination of encorafenib, a potent BRAF inhibitor, and binimetinib, a potent MEK inhibitor, demonstrated durable antitumor activity with a manageable safety profile in this patient population. On the basis of the results of this study, the combination of encorafenib plus binimetinib was approved by the US Food and Drug Administration on October 11, 2023, for patients with BRAF V600E-mutant metastatic NSCLC. In this review, we summarize the efficacy and safety of encorafenib plus binimetinib from the PHAROS study. In addition, we discuss strategies to manage adverse reactions with this combination therapy with the intent of minimizing unnecessary treatment discontinuations in these patients.
Collapse
Affiliation(s)
- Christina Baik
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael L Cheng
- University of California, San Francisco, San Francisco, CA, USA
| | - Martin Dietrich
- US Oncology and University of Central Florida, Orlando, FL, USA
| | - Jhanelle E Gray
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nagla A Karim
- Inova Schar Cancer Institute, University of Virginia, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA.
| |
Collapse
|
84
|
Kim M, Shim HS, Kim S, Lee IH, Kim J, Yoon S, Kim HD, Park I, Jeong JH, Yoo C, Cheon J, Kim IH, Lee J, Hong SH, Park S, Jung HA, Kim JW, Kim HJ, Cha Y, Lim SM, Kim HS, Lee CK, Kim JH, Chun SH, Yun J, Park SY, Lee HS, Cho YM, Nam SJ, Na K, Yoon SO, Lee A, Jang KT, Yun H, Lee S, Kim JH, Kim WS. Clinical Practice Recommendations for the Use of Next-Generation Sequencing in Patients with Solid Cancer: A Joint Report from KSMO and KSP. Cancer Res Treat 2024; 56:721-742. [PMID: 38037319 PMCID: PMC11261187 DOI: 10.4143/crt.2023.1043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
In recent years, next-generation sequencing (NGS)-based genetic testing has become crucial in cancer care. While its primary objective is to identify actionable genetic alterations to guide treatment decisions, its scope has broadened to encompass aiding in pathological diagnosis and exploring resistance mechanisms. With the ongoing expansion in NGS application and reliance, a compelling necessity arises for expert consensus on its application in solid cancers. To address this demand, the forthcoming recommendations not only provide pragmatic guidance for the clinical use of NGS but also systematically classify actionable genes based on specific cancer types. Additionally, these recommendations will incorporate expert perspectives on crucial biomarkers, ensuring informed decisions regarding circulating tumor DNA panel testing.
Collapse
Affiliation(s)
- Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - In Hee Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung-Don Kim
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaekyung Cheon
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sook Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Han Jo Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Yongjun Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Choong-kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jina Yun
- Division of Hematology/Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
85
|
Li Z, Lu W, Yin F, Huang A. YBX1 as a prognostic biomarker and potential therapeutic target in hepatocellular carcinoma: A comprehensive investigation through bioinformatics analysis and in vitro study. Transl Oncol 2024; 45:101965. [PMID: 38688048 PMCID: PMC11070759 DOI: 10.1016/j.tranon.2024.101965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/08/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUNDS Y-box binding protein 1 (YBX1) is a DNA/RNA binding protein known to contribute to the progression of various malignancies, however, a comprehensive pan-cancer analysis to investigate YBX1 across a broad spectrum of cancer types has not yet been conducted. METHODS We utilized the TIMER database for a comprehensive pan-cancer analysis and assessed YBX-1 expression via the TCGA and GEO databases. The relationship between YBX-1 expression and tumor-infiltrating cells was examined using TIMER and the R programming language. To evaluate the prognostic value of YBX1, we performed Kaplan-Meier plots and Cox regression analyses. Through LinkedOmics, we identified genes significantly correlated with YBX-1. The WEB-based Gene SeT AnaLysis Toolkit was used for KEGG pathway enrichment analysis. Additionally, using shRNA-mediated knockdown, we explored the potential role of YBX1 in tumor cell biology. RESULTS Our study identifies pronounced overexpression of YBX-1 across multiple cancer types, correlating with adverse outcomes, notably in liver hepatocellular carcinoma (LIHC). A distinct association between elevated YBX-1 expression and heightened immune cell infiltration suggests YBX-1's potential role in reshaping the tumor microenvironment. Intriguingly, our KEGG pathway analysis indicated a tight nexus between YBX-1 expression and lipid metabolism. Moreover, the suppression of YBX-1 via shRNA revealed diminished cellular proliferation and marked reductions in crucial molecules steering the fatty acid synthesis pathway, implicating YBX-1's potential regulatory role in lipid metabolism within LIHC. CONCLUSIONS YBX-1 serves as a favorable prognostic indicator in various cancers, particularly in liver hepatocellular carcinoma. Targeting YBX1 in HCC offers potential therapeutic strategies. This work paves the way for fresh insights into targeted therapeutic approaches for cancers, especially benefiting liver hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Zizhen Li
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou 510000, China
| | - Feng Yin
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou 510000, China
| | - Amin Huang
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510000, China.
| |
Collapse
|
86
|
Di Leo L, Pagliuca C, Kishk A, Rizza S, Tsiavou C, Pecorari C, Dahl C, Pacheco MP, Tholstrup R, Brewer JR, Berico P, Hernando E, Cecconi F, Ballotti R, Bertolotto C, Filomeni G, Gjerstorff MF, Sauter T, Lovat P, Guldberg P, De Zio D. AMBRA1 levels predict resistance to MAPK inhibitors in melanoma. Proc Natl Acad Sci U S A 2024; 121:e2400566121. [PMID: 38870061 PMCID: PMC11194594 DOI: 10.1073/pnas.2400566121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Intrinsic and acquired resistance to mitogen-activated protein kinase inhibitors (MAPKi) in melanoma remains a major therapeutic challenge. Here, we show that the clinical development of resistance to MAPKi is associated with reduced tumor expression of the melanoma suppressor Autophagy and Beclin 1 Regulator 1 (AMBRA1) and that lower expression levels of AMBRA1 predict a poor response to MAPKi treatment. Functional analyses show that loss of AMBRA1 induces phenotype switching and orchestrates an extracellular signal-regulated kinase (ERK)-independent resistance mechanism by activating focal adhesion kinase 1 (FAK1). In both in vitro and in vivo settings, melanomas with low AMBRA1 expression exhibit intrinsic resistance to MAPKi therapy but higher sensitivity to FAK1 inhibition. Finally, we show that the rapid development of resistance in initially MAPKi-sensitive melanomas can be attributed to preexisting subclones characterized by low AMBRA1 expression and that cotreatment with MAPKi and FAK1 inhibitors (FAKi) effectively prevents the development of resistance in these tumors. In summary, our findings underscore the value of AMBRA1 expression for predicting melanoma response to MAPKi and supporting the therapeutic efficacy of FAKi to overcome MAPKi-induced resistance.
Collapse
Affiliation(s)
- Luca Di Leo
- Melanoma Research Team, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Chiara Pagliuca
- Melanoma Research Team, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Ali Kishk
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux4365, Luxembourg
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Christina Tsiavou
- Melanoma Research Team, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Chiara Pecorari
- Redox Biology Group, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Christina Dahl
- Molecular Diagnostics Group, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Maria Pires Pacheco
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux4365, Luxembourg
| | - Rikke Tholstrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense5230, Denmark
| | - Jonathan Richard Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense5230, Denmark
| | - Pietro Berico
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Eva Hernando
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Francesco Cecconi
- Cell Stress and Survival, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen2100, Denmark
- Faculty of Medicine and Surgery, Università Cattolica del “Sacro Cuore”, Fondazione Policlinico Gemelli—Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome00136, Italy
| | - Robert Ballotti
- Université Côte d’Azur, Nice06200, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, Centre Méditerranéen de Médecine Moléculaire, Nice06200, France
| | - Corine Bertolotto
- Université Côte d’Azur, Nice06200, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020, Centre Méditerranéen de Médecine Moléculaire, Nice06200, France
| | - Giuseppe Filomeni
- Redox Biology Group, Danish Cancer Institute, Copenhagen2100, Denmark
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense5230, Denmark
- Department of Oncology, Odense University Hospital, Odense5000, Denmark
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux4365, Luxembourg
| | - Penny Lovat
- Translational and Clinical Research Institute, Medical School, Newcastle University, Newcastle upon TyneNE2 4HH, United Kingdom
| | - Per Guldberg
- Molecular Diagnostics Group, Danish Cancer Institute, Copenhagen2100, Denmark
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense5230, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen2100, Denmark
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense5230, Denmark
| |
Collapse
|
87
|
Slominski RM, Kim TK, Janjetovic Z, Brożyna AA, Podgorska E, Dixon KM, Mason RS, Tuckey RC, Sharma R, Crossman DK, Elmets C, Raman C, Jetten AM, Indra AK, Slominski AT. Malignant Melanoma: An Overview, New Perspectives, and Vitamin D Signaling. Cancers (Basel) 2024; 16:2262. [PMID: 38927967 PMCID: PMC11201527 DOI: 10.3390/cancers16122262] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Melanoma, originating through malignant transformation of melanin-producing melanocytes, is a formidable malignancy, characterized by local invasiveness, recurrence, early metastasis, resistance to therapy, and a high mortality rate. This review discusses etiologic and risk factors for melanoma, diagnostic and prognostic tools, including recent advances in molecular biology, omics, and bioinformatics, and provides an overview of its therapy. Since the incidence of melanoma is rising and mortality remains unacceptably high, we discuss its inherent properties, including melanogenesis, that make this disease resilient to treatment and propose to use AI to solve the above complex and multidimensional problems. We provide an overview on vitamin D and its anticancerogenic properties, and report recent advances in this field that can provide solutions for the prevention and/or therapy of melanoma. Experimental papers and clinicopathological studies on the role of vitamin D status and signaling pathways initiated by its active metabolites in melanoma prognosis and therapy are reviewed. We conclude that vitamin D signaling, defined by specific nuclear receptors and selective activation by specific vitamin D hydroxyderivatives, can provide a benefit for new or existing therapeutic approaches. We propose to target vitamin D signaling with the use of computational biology and AI tools to provide a solution to the melanoma problem.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Department of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Tae-Kang Kim
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Zorica Janjetovic
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Ewa Podgorska
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Katie M. Dixon
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Rebecca S. Mason
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia; (K.M.D.); (R.S.M.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia;
| | - Rahul Sharma
- Department of Biomedical Informatics and Data Science, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - David K. Crossman
- Department of Genetics and Bioinformatics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Craig Elmets
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Chander Raman
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
| | - Anton M. Jetten
- Cell Biology Section, NIEHS—National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrzej T. Slominski
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.-K.K.); (Z.J.); (E.P.); (C.E.); (C.R.)
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, Veteran Administration Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
88
|
Grabbe P, Borchers MS, Gschwendtner KM, Strobel S, Wild B, Kirchner M, Kälber K, Rendon A, Steininger J, Meier F, Hassel JC, Bieber C. An Online Decision Aid for Patients With Metastatic Melanoma—Results of the Randomized Controlled Trial “PEF-Immun”. DEUTSCHES ARZTEBLATT INTERNATIONAL 2024; 121:385-392. [PMID: 38566437 PMCID: PMC11460262 DOI: 10.3238/arztebl.m2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Treatment decisions in metastatic melanoma (MM) are highly dependent on patient preferences and require the patients' involvement. The complexity of treatment options with their individual advantages and disadvantages is often overwhelming. We therefore developed an online patient decision aid (PtDA) to facilitate shared decision making (SDM). METHODS To evaluate the PtDA we conducted a two-armed, twocenter, prospective, open randomized controlled trial with MM patients who were facing a decision about first-line treatment. The patients were allotted randomly in a 1:1 ratio to an intervention group (IG) with access to the PtDA before discussion with a physician or to a control group (CG) without access to the PtDA. The primary endpoint was knowledge about the options for first-line treatment (multiple-choice test, 10 items, range 0-40 points). The secondary endpoints were the SDM (third-party ratings of audio recordings of the treatment discussions) and satisfaction with the decision at the follow-up visit. RESULTS Of the 128 randomized patients, 120 completed the baseline questionnaire and were analyzed (59% male, median age 66 years). The primary endpoint, i.e., the mean difference in knowledge after discussion with a physician, differed significantly between the IG and the CG (-3.22, 95% CI [-6.32; -0.12], p = 0.042). No differences were found for the secondary endpoints, SDM and satisfaction with the decision. The patients in the IG rated the PtDA as very useful. CONCLUSION The PtDA improved the knowledge of patients with MM about the options for treatment. Both groups were highly satisfied with their treatment decisions. However, additional physician training seems necessary to promote SDM.
Collapse
Affiliation(s)
- Pia Grabbe
- *Joint first authors
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Milena S. Borchers
- *Joint first authors
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Kathrin M. Gschwendtner
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Sophia Strobel
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Wild
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Marietta Kirchner
- Institute of Medical Biometry, Heidelberg University Hospital, Heidelberg
| | - Katharina Kälber
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Adriana Rendon
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Steininger
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at the Technical University Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at the Technical University Dresden, Dresden, Germany
| | - Jessica C. Hassel
- *Joint last authors
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | | |
Collapse
|
89
|
Li KH, Cheung PCF, Petrella TM, Zhang L, Poon IDT, Menjak IB. Clinical outcomes of multisite moderate to high dose radiotherapy for patients with metastatic melanoma. PRECISION RADIATION ONCOLOGY 2024; 8:62-69. [PMID: 40336647 PMCID: PMC11934978 DOI: 10.1002/pro6.1224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/24/2024] [Accepted: 02/22/2024] [Indexed: 05/09/2025] Open
Abstract
Background and purpose This study aimed to summarize the clinical outcomes of patients with metastatic melanoma who received high-dose radiation prior to or during systemic therapy at a single academic institution. Methods We identified patients with metastatic melanoma who underwent high-dose radiation therapy (HDRT) for extracranial metastases prior to or during systemic therapy from 2010 to 2018. Treatment indications included oligometastases, oligoprogression, and local control. Using the Kaplan-Meier method, we plotted overall survival (OS), progression-free survival-1 (PFS1), and PFS2. Competing risk analysis determined the cumulative incidence of local failure (LF) and the time to start or change systemic therapy (SCST). Univariate and multivariable analyses were used to identify predictive factors. Results We analyzed 34 patients with 79 lesions, with a median follow-up of 17.4 months. Sixty-eight percent of patients received systemic therapy after the first HDRT. The median OS was 22 months, with brain metastases before HDRT being a significant predictor in multivariable analysis. The median PFS1 for first-line HDRT was 4.1 months, and the median PFS2 was 3.9 months. Rates of LF were 10.3% at 12 months and 11.7% at 24 months. The incidence of SCST following HDRT was 59.8% at 12 months and 76.1% at 24 months, with radiation targeted at the lung associated with a lower incidence of SCST. Conclusion HDRT for treating metastatic lesions in melanoma demonstrated excellent local control and may play a role in delaying SCST. Additional courses of HDRT may provide cumulative benefits.
Collapse
Affiliation(s)
- Kelly H. Li
- Department of Medical OncologyBC Cancer AgencyVancouverCanada
| | - Patrick CF. Cheung
- Department of Radiation OncologyOdette Cancer Centre, Toronto, Sunnybrook Health Sciences CentreTorontoCanada
| | - Teresa M. Petrella
- Department of Medical OncologyOdette Cancer Centre, Sunnybrook Health Sciences CentreTorontoCanada
| | | | - Ian DT. Poon
- Department of Radiation OncologyOdette Cancer Centre, Toronto, Sunnybrook Health Sciences CentreTorontoCanada
| | - Ines B. Menjak
- Department of Medical OncologyOdette Cancer Centre, Sunnybrook Health Sciences CentreTorontoCanada
| |
Collapse
|
90
|
Trouiller JB, Nikolaidis GF, Macabeo B, Meyer N, Gerlier L, Schlueter M, Laramee P. Cost-effectiveness of encorafenib with binimetinib in unresectable or metastatic BRAF-mutant melanoma. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2024; 25:641-653. [PMID: 37433888 DOI: 10.1007/s10198-023-01614-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 06/19/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVE The objective of this study was to determine the cost-effectiveness of encorafenib with binimetinib (EncoBini) as compared to other targeted double combination therapies, namely dabrafenib with trametinib (DabraTrame) and vemurafenib with cobimetinib (VemuCobi), for the treatment of BRAF V600-mutant unresectable or metastatic melanoma (MM) from the French payer perspective. METHODS A partitioned survival model was developed considering a lifetime horizon. The model structure simulated the clinical pathway of patients with BRAF V600-mutant MM. Clinical effectiveness and safety inputs were sourced from the COLUMBUS trial, a network meta-analysis and published literature. Costs, resource use, and the quality of life inputs were obtained from the literature and appropriate French sources. RESULTS Over a lifetime horizon, EncoBini was associated, on average, with reduced costs and increased quality adjusted life years (QALYs), dominating both targeted double combination therapies. For a willingness-to-pay threshold of €90,000 per QALY, the probability of EncoBini being cost-effective against either comparator remained above 80%. The most influential model parameters were the hazard ratios for the overall survival of EncoBini vs DabraTrame and VemuCobi, the pre- and post-progression utility values, as well as treatment dosages and the relative dose intensity of all interventions. CONCLUSION EncoBini is associated with reduced costs and increased QALYs, dominating other targeted double combination therapies (DabraTrame, VemuCobi) for patients with BRAF V600-mutant MM in France. EncoBini is a highly cost-effective intervention in MM.
Collapse
Affiliation(s)
- Jean-Baptiste Trouiller
- Pierre Fabre Laboratories, 33 avenue Emile Zola, 92100, Boulogne-Billancourt, France.
- Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France.
| | | | - Bérengère Macabeo
- Pierre Fabre Laboratories, 33 avenue Emile Zola, 92100, Boulogne-Billancourt, France
- Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
| | - Nicolas Meyer
- Cabinet médical, Clinique Médipôle Garonne, 45 rue de Gironis, 31067, Toulouse Cedex 1, France
| | | | | | - Philippe Laramee
- Pierre Fabre Laboratories, 33 avenue Emile Zola, 92100, Boulogne-Billancourt, France
- Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
| |
Collapse
|
91
|
Naci H, Zhang Y, Woloshin S, Guan X, Xu Z, Wagner AK. Overall survival benefits of cancer drugs initially approved by the US Food and Drug Administration on the basis of immature survival data: a retrospective analysis. Lancet Oncol 2024; 25:760-769. [PMID: 38754451 DOI: 10.1016/s1470-2045(24)00152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/24/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND New cancer drugs can be approved by the US Food and Drug Administration (FDA) on the basis of surrogate endpoints while data on overall survival are still incomplete or immature, with too few deaths for meaningful analysis. We aimed to evaluate whether clinical trials with immature survival data generated evidence of overall survival benefit during the period after marketing authorisation, and where that evidence was reported. METHODS In this retrospective analysis, we searched Drugs@FDA to identify cancer drug indications approved between Jan 1, 2001, and Dec 31, 2018, on the basis of immature survival data. We systematically collected publicly available data on postapproval overall survival results in labelling (Drugs@FDA), journal publications (MEDLINE via PubMed), and clinical trial registries (ClinicalTrials.gov). The primary outcome was availability of statistically significant overall survival benefits during the period after marketing authorisation (until March 31, 2023). Additionally, we evaluated the availability and timing of overall survival findings in labelling, journal publications, and ClinicalTrials.gov records. FINDINGS During the study period, the FDA granted marketing authorisation to 223 cancer drug indications, 95 of which had overall survival as an endpoint. 39 (41%) of these 95 indications had immature survival data. After a minimum of 4·3 years of follow-up during the period after marketing authorisation (and median 8·2 years [IQR 5·3-12·0] since FDA approval), additional survival data from the pivotal trials became available in either revised labelling or publications, or both, for 38 (97%) of 39 indications. Additional data on overall survival showed a statistically significant benefit in 12 (32%) of 38 indications, whereas mature data yielded statistically non-significant overall survival findings for 24 (63%) indications. Statistically significant evidence of overall survival benefit was reported in either labelling or publications a median of 1·5 years (IQR 0·8-2·3) after initial approval. The median time to availability of statistically non-significant overall survival results was 3·3 years (2·2-4·5). The availability of overall survival results on ClinicalTrials.gov varied considerably. INTERPRETATION Fewer than a third of indications approved with immature survival data showed a statistically significant overall survival benefit after approval. Notable inconsistencies in timing and availability of information after approval across different sources emphasise the need for better reporting standards. FUNDING None.
Collapse
Affiliation(s)
- Huseyin Naci
- Department of Health Policy, London School of Economics and Political Science, London, UK; The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA.
| | - Yichen Zhang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Steven Woloshin
- The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA; The Center for Medicine in the Media, Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Xiaodong Guan
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ziyue Xu
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Anita K Wagner
- The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA; Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| |
Collapse
|
92
|
Dima D, Lopetegui‐Lia N, Ogbue O, Osantowski B, Ullah F, Jia X, Song JM, Gastman B, Isaacs J, Kennedy LB, Funchain P. Real-world outcomes of patients with resected stage III melanoma treated with adjuvant therapies. Cancer Med 2024; 13:e7257. [PMID: 39031560 PMCID: PMC11190025 DOI: 10.1002/cam4.7257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/28/2023] [Accepted: 04/27/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Both immunotherapy (IO) and targeted therapy (TT) are used as adjuvant (adj) treatment for stage III melanoma, however, data describing real-world outcomes are limited. In addition, a significant proportion of patients relapse, for whom best management is unclear. The aim of our study was to assess the efficacy, and safety of adj anti-PD1 IO and TT in a real-world cohort of patients with resected stage III melanoma, and further delineate patterns of recurrence and treatment strategies. METHODS We retrospectively analyzed 130 patients who received adj therapy (100 anti-PD1 IO and 30 TT). RESULTS At a median follow-up of 30 months, median relapse-free survival (RFS) was 24.6 (95% CI, 17-not reached [NR]) versus 64 (95% CI, 29.5-NR) months for the TT and IO groups, respectively (p = 0.26). Median overall survival (OS) was NR for either subgroup. At data cutoff, 77% and 82% of patients in TT and IO arms were alive. A higher number of grade ≥3 treatment-related adverse events (AEs) were noted in the IO group (11% vs. 3%), however, a higher proportion of patients permanently discontinued adj therapy in the TT group (43% vs. 11%) due to toxicity. Strategies at relapse and outcomes were variable based on location and timing of recurrence. A significant number of patients who relapsed after adj IO received a second round of IO. Among them, patients who were off adj IO at relapse had superior second median RFS (mRFS2), compared to those who relapsed while on adj IO; mRFS2 was NR versus 5.1 months (95% CI, 2.5-NR), respectively, p = 0.02. CONCLUSION In summary, both TT and IO yielded prolonged RFS in a real-world setting, however, longer follow-up is needed to determine any potential OS benefit. Adj therapy, particularly TT, may not be as well tolerated as suggested in clinical trials, with lower completion rates (59% vs. 74%) in a real-life setting. Overall, patients who relapse during adj therapy have poor outcomes, while patients who relapse after discontinuation of adj IO therapy appear to benefit from IO re-treatment.
Collapse
Affiliation(s)
- Danai Dima
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Nerea Lopetegui‐Lia
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Olisaemeka Ogbue
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Bennett Osantowski
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Fauzia Ullah
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Xuefei Jia
- Department of BiostatisticsCleveland Clinic FoundationClevelandOhioUSA
| | - Jung Min Song
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Brian Gastman
- Department of Plastic SurgeryCleveland Clinic FoundationClevelandOhioUSA
| | - James Isaacs
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Lucy Boyce Kennedy
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
| | - Pauline Funchain
- Department of Hematology‐OncologyTaussig Cancer Institute, Cleveland Clinic FoundationClevelandOhioUSA
- Division of Oncology, Stanford Cancer InstituteStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
93
|
Hennemann A, Puzenat E, Decreuse M, Vuillier F, Nardin C, Aubin F. Intracranial hemorrhage caused by dabrafenib and trametinib therapy for metastatic melanoma. Melanoma Res 2024; 34:280-282. [PMID: 38602773 DOI: 10.1097/cmr.0000000000000820] [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: 04/13/2024]
Abstract
Although generally well tolerated compared with chemotherapy, molecular targeted therapy used in metastatic melanoma may be associated with life-threatening toxicity. We report the case of a patient with metastatic melanoma treated by dabrafenib plus trametinib who developed intracranial hemorrhage. Physicians should be aware of this rare but life-threatening adverse event of B-rapidly accelerated fibrosarcoma (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors. However, they should be also careful about the bleeding origin, which can prove to be a new onset of melanoma metastasis or anticoagulation overdose, or even an uncontrolled arterial hypertension.
Collapse
Affiliation(s)
| | - Eve Puzenat
- Department of Dermatology, University Hospital, Inserm 1098
| | - Marion Decreuse
- Department of Neurology, University Hospital, Besançon, France
| | | | - Charlée Nardin
- Department of Dermatology, University Hospital, Inserm 1098
| | - François Aubin
- Department of Dermatology, University Hospital, Inserm 1098
| |
Collapse
|
94
|
Costa Svedman F, Liapi M, Månsson-Broberg A, Chatzidionysiou K, Egyhazi Brage S. Effect of glucocorticoids for the management of immune-related adverse events on outcome in melanoma patients treated with immunotherapy-a retrospective and biomarker study. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 22:100713. [PMID: 38952418 PMCID: PMC11215956 DOI: 10.1016/j.iotech.2024.100713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Background Immune-related adverse events (IRAEs) during therapy with immune checkpoint inhibitors (ICIs) are common, and their management sometimes requires glucocorticoids (GCs). Predictors for development of IRAEs and data about the impact of GCs on clinical outcome are missing. We evaluated the impact of GCs to treat IRAEs on clinical outcome, and plasmatic inflammatory proteins as predictors for IRAEs. Patients and methods Patients with melanoma (n = 98) treated with ICIs at Karolinska University Hospital were included. Clinical information and data regarding prescription of systemic GCs were collected. Baseline plasma samples (n = 57) were analyzed for expression of 92 inflammatory proteins. Results Forty-four patients developed at least one IRAE requiring systemic GCs and the most common was hypocortisolemia (n = 11). A median overall survival of 72.8 months for patients developing IRAEs requiring GCs, 17.7 months for those who did not, and 1.4 months for individuals receiving GCs at baseline was observed in Kaplan-Meier curves (P = 0.001). In immortal time bias adjusted analysis, patients receiving steroids to treat IRAE survived slightly longer, even though this time trend was not statistically significant. The median overall survival was 29 months for those treated with GCs within 60 days after ICIs start and was not reached for patients receiving GCs later. The number of ICI cycles was higher in subjects receiving GCs after 60 days (P = 0.0053). Hypocortisolemia occurred mainly in males (10/11) and correlated with favorable outcome. Male patients with hypocortisolemia had lower expression of interleukin 8, transforming growth factor-α, and fibroblast growth factor 5 and higher expression of Delta/Notch-like epidermal growth factor-related receptor. Conclusions GCs may be used to treat IRAEs without major concern. GCs early during ICIs may, however, impact clinical outcome negatively. The prognostic value of hypocortisolemia and inflammation proteins as biomarkers should be further investigated.
Collapse
Affiliation(s)
- F. Costa Svedman
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - M. Liapi
- Department of Rheumatology, Theme Inflammation and Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - A. Månsson-Broberg
- Theme Heart and Vascular, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - K. Chatzidionysiou
- Department of Rheumatology, Theme Inflammation and Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - S. Egyhazi Brage
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
95
|
Hassel JC, Zimmer L. [Side effects of dermato-oncologic therapies]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:466-475. [PMID: 38802653 DOI: 10.1007/s00105-024-05354-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/12/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) such as PD(L)1 and CTLA4 antibodies as well as targeted therapies such as BRAF and MEK inhibitors have significantly improved the systemic treatment of skin cancer in adjuvant and advanced therapy settings. All these drugs differ in their spectrum of side effects. MATERIALS AND METHODS The aim of this article is to provide an overview of the spectrum of side effects of dermato-oncological therapies and their management, taking into account the current literature. RESULTS The most important side effects of ICIs, the CCR4 inhibitor mogamulizumab, the ImmTAC tebentafusp, the BRAF and MEK inhibitors and the multityrosine kinase inhibitor imatinib are considered. CONCLUSIONS Side effects can manifest themselves in all organ systems. Chronic side effects and long-term harm are possible, especially with ICIs, and require close therapy monitoring and patient education. Knowledge of the side effects and the temporal, sometimes delayed course of their occurrence are essential for diagnosis and prompt initiation of therapy.
Collapse
Affiliation(s)
- Jessica C Hassel
- Medizinische Fakultät, Hautklinik und Nationales Centrum für Tumorerkrankungen (NCT), NCT Heidelberg, eine Partnerschaft zwischen DKFZ und dem Universitätsklinikum Heidelberg, Universität Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Deutschland.
| | - Lisa Zimmer
- Klinik für Dermatologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| |
Collapse
|
96
|
Scardaci R, Berlinska E, Scaparone P, Vietti Michelina S, Garbo E, Novello S, Santamaria D, Ambrogio C. Novel RAF-directed approaches to overcome current clinical limits and block the RAS/RAF node. Mol Oncol 2024; 18:1355-1377. [PMID: 38362705 PMCID: PMC11161739 DOI: 10.1002/1878-0261.13605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/30/2023] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Mutations in the RAS-RAF-MEK-ERK pathway are frequent alterations in cancer and RASopathies, and while RAS oncogene activation alone affects 19% of all patients and accounts for approximately 3.4 million new cases every year, less frequent alterations in the cascade's downstream effectors are also involved in cancer etiology. RAS proteins initiate the signaling cascade by promoting the dimerization of RAF kinases, which can act as oncoproteins as well: BRAFV600E is the most common oncogenic driver, mutated in the 8% of all malignancies. Research in this field led to the development of drugs that target the BRAFV600-like mutations (Class I), which are now utilized in clinics, but cause paradoxical activation of the pathway and resistance development. Furthermore, they are ineffective against non-BRAFV600E malignancies that dimerize and could be either RTK/RAS independent or dependent (Class II and III, respectively), which are still lacking an effective treatment. This review discusses the recent advances in anti-RAF therapies, including paradox breakers, dimer-inhibitors, immunotherapies, and other novel approaches, critically evaluating their efficacy in overcoming the therapeutic limitations, and their putative role in blocking the RAS pathway.
Collapse
Affiliation(s)
- Rossella Scardaci
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ewa Berlinska
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Pietro Scaparone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Sandra Vietti Michelina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Edoardo Garbo
- Department of OncologyUniversity of Torino, San Luigi HospitalOrbassanoItaly
| | - Silvia Novello
- Department of OncologyUniversity of Torino, San Luigi HospitalOrbassanoItaly
| | - David Santamaria
- Centro de Investigación del CáncerCSIC‐Universidad de SalamancaSpain
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| |
Collapse
|
97
|
Schadendorf D, Dummer R, Flaherty KT, Robert C, Arance A, de Groot JWB, Garbe C, Gogas HJ, Gutzmer R, Krajsová I, Liszkay G, Loquai C, Mandalà M, Yamazaki N, Queirolo P, Guenzel C, Polli A, Thakur M, di Pietro A, Ascierto PA. COLUMBUS 7-year update: A randomized, open-label, phase III trial of encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF V600E/K-mutant melanoma. Eur J Cancer 2024; 204:114073. [PMID: 38723373 DOI: 10.1016/j.ejca.2024.114073] [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: 02/03/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Treatment with encorafenib plus binimetinib and encorafenib monotherapy is associated with improved progression-free survival (PFS) and overall survival (OS) compared with vemurafenib in patients with BRAF V600E/K-mutant metastatic melanoma. We report results from the 7-year analysis of COLUMBUS part 1 (NCT01909453) at 99.7 months (median duration between randomization and data cutoff). METHODS 577 patients with locally advanced unresectable or metastatic BRAF V600E/K-mutant melanoma who were treatment-naive or progressed after first-line immunotherapy were randomized 1:1:1 to encorafenib 450 mg once daily (QD) plus binimetinib 45 mg twice daily (BID) (n = 192), vemurafenib 960 mg BID (n = 191), or encorafenib monotherapy 300 mg QD (n = 194). No prior BRAF/MEK inhibitor was allowed. RESULTS Seven-year PFS and OS rates (95 % CI) were 21.2 % (14.7-28.4 %) and 27.4 % (21.2-33.9%) in the encorafenib plus binimetinib arm and 6.4 % (2.1-14.0 %) and 18.2 % (12.8-24.3 %) in the vemurafenib arm, respectively. Median melanoma-specific survival (95 % CI) was 36.8 months (27.7-51.5 months) in the encorafenib plus binimetinib arm and 19.3 months (14.8-25.9 months) in the vemurafenib arm. Thirty-four long-term responders (complete/partial response ongoing at 7 years) were identified across arms. CONCLUSIONS This is the longest follow-up from a phase III trial of BRAF/MEK inhibitor combination in BRAF V600E/K-mutant metastatic melanoma. Safety results were consistent with the known tolerability profile of encorafenib plus binimetinib. Results support the long-term efficacy and known safety of encorafenib plus binimetinib in this population and provide new insights on long-term responders. Interactive data visualization is available at the COLUMBUS dashboard (https://clinical-trials.dimensions.ai/columbus7/).
Collapse
Affiliation(s)
- Dirk Schadendorf
- University Hospital Essen, West German Cancer Center and German Cancer Consortium, Partner Site Essen, Essen, Germany; National Center for Tumor Diseases West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University of Duisburg-Essen, Essen, Germany.
| | | | | | - Caroline Robert
- Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Ana Arance
- Hospital Clinic of Barcelona and Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Claus Garbe
- University Hospital Tübingen, Tübingen, Germany
| | - Helen J Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | - Ralf Gutzmer
- Hannover Medical School, Hannover, and Ruhr-University Bochum, Minden Campus, Germany
| | | | | | - Carmen Loquai
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | - Paola Queirolo
- Melanoma and Sarcoma Division, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | | | | | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| |
Collapse
|
98
|
Rozendorn N, Shutan I, Feinmesser G, Grynberg S, Hodadov H, Alon E, Asher N. Real-World Outcomes of Inoperable and Metastatic Cutaneous Head and Neck Melanoma Patients. Laryngoscope 2024; 134:2762-2770. [PMID: 38230960 DOI: 10.1002/lary.31290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
OBJECTIVE This study aims to describe the overall survival (OS) and to identify associated prognostic factors in patients with inoperable and metastatic cutaneous melanoma of the head and neck (H&N) region, undergoing modern systemic treatments. METHODS This is a retrospective single institutional study. Data on all consecutive H&N melanoma patients treated with systemic oncologic treatments between 2015 and 2022 were collected from electronic medical files. Kaplan-Meier curves were used to describe survival and Cox regression analysis was used to identify patient and tumor factors associated with prognosis. RESULTS A total of 144 patients were included. Median OS was 45 months (95% confidence interval [CI] 28-65 m). On univariable analysis for OS, the primary disease site, specifically the nape and neck (hazard ratio [HR] 3.3, 95% CI 1.4-7.7, p = 0.007), high Eastern Cooperative Oncology Group Performance Status ([ECOG-PS], HR 2.5, 95% CI = 1.9-3.3, p < 0.001), high lactate dehydrogenase (LDH) levels (HR 2.8, 95% CI = 1.7-4.6, p < 0.001), and treatment with targeted therapy (TT) as compared with immunotherapy (HR 2.6, 95% CI = 1.06-6.3, p = 0.03) were all associated with shorter OS. High-grade adverse events (AEs) were associated with a longer OS (HR 0.41, 95% CI = 0.25-0.68, p = 0.001). On multivariable analysis for OS, the ECOG-PS, LDH levels, site of disease, and the development of moderate-severe AEs remained significant. CONCLUSIONS In the era of modern oncologic treatments, the prognosis of inoperable and metastatic cutaneous H&N melanoma aligns with other cutaneous melanomas. Primary tumor site of the nape and neck region emerges as a significant prognostic factor. LEVEL OF EVIDENCE 3 Laryngoscope, 134:2762-2770, 2024.
Collapse
Affiliation(s)
- Noa Rozendorn
- Department of Otolaryngology-Head and Neck Surgery, Sheba Medica Center, Ramat Gan, Israel
| | - Itay Shutan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Feinmesser
- Department of Otolaryngology-Head and Neck Surgery, Sheba Medica Center, Ramat Gan, Israel
| | - Shirly Grynberg
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Hadas Hodadov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Alon
- Department of Otolaryngology-Head and Neck Surgery, Sheba Medica Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nethanel Asher
- Skin Cancer and Melanoma Center, Davidoff Center, Beilinson Medical Center, Petah Tikva, Israel
| |
Collapse
|
99
|
Rasmussen DM, Semonis MM, Greene JT, Muretta JM, Thompson AR, Toledo Ramos S, Thomas DD, Pomerantz WCK, Freedman TS, Levinson NM. Allosteric coupling asymmetry mediates paradoxical activation of BRAF by type II inhibitors. eLife 2024; 13:RP95481. [PMID: 38742856 PMCID: PMC11093583 DOI: 10.7554/elife.95481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
The type II class of RAF inhibitors currently in clinical trials paradoxically activate BRAF at subsaturating concentrations. Activation is mediated by induction of BRAF dimers, but why activation rather than inhibition occurs remains unclear. Using biophysical methods tracking BRAF dimerization and conformation, we built an allosteric model of inhibitor-induced dimerization that resolves the allosteric contributions of inhibitor binding to the two active sites of the dimer, revealing key differences between type I and type II RAF inhibitors. For type II inhibitors the allosteric coupling between inhibitor binding and BRAF dimerization is distributed asymmetrically across the two dimer binding sites, with binding to the first site dominating the allostery. This asymmetry results in efficient and selective induction of dimers with one inhibited and one catalytically active subunit. Our allosteric models quantitatively account for paradoxical activation data measured for 11 RAF inhibitors. Unlike type II inhibitors, type I inhibitors lack allosteric asymmetry and do not activate BRAF homodimers. Finally, NMR data reveal that BRAF homodimers are dynamically asymmetric with only one of the subunits locked in the active αC-in state. This provides a structural mechanism for how binding of only a single αC-in inhibitor molecule can induce potent BRAF dimerization and activation.
Collapse
Affiliation(s)
- Damien M Rasmussen
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Manny M Semonis
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
| | - Joseph T Greene
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
| | - Joseph M Muretta
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Andrew R Thompson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | | | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | | | - Tanya S Freedman
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
- Center for Immunology, University of MinnesotaMinneapolisUnited States
- Masonic Cancer Center, University of MinnesotaMinneapolisUnited States
| | - Nicholas M Levinson
- Department of Pharmacology, University of MinnesotaMinneapolisUnited States
- Masonic Cancer Center, University of MinnesotaMinneapolisUnited States
| |
Collapse
|
100
|
Adamopoulos C, Papavassiliou KA, Poulikakos PI, Papavassiliou AG. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:4633. [PMID: 38731852 PMCID: PMC11083651 DOI: 10.3390/ijms25094633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib-trametinib, in 2017, and encorafenib-binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|