1
|
Amrane K, Le Noac'h P, Hemon P, Abgral R, Le Meur C, Pradier O, Misery L, Legoupil D, Berthou C, Uguen A. MHC class II: a predictor of outcome in melanoma treated with immune checkpoint inhibitors. Melanoma Res 2025; 35:176-186. [PMID: 39945603 DOI: 10.1097/cmr.0000000000001022] [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: 05/03/2025]
Abstract
This study aimed to evaluate the predictive value of MHC class II (MHC-II) expression by melanoma cells in a large cohort of metastatic cutaneous melanoma patients treated with immune checkpoint inhibitors (ICIs). We conducted a single-center, retrospective study involving stage IV cutaneous melanoma patients who received ICI as first-line therapy. MHC-II expression in melanoma cells was quantified using dual-color anti-SOX10 and anti-MHC-II immunohistochemistry on tumor samples from 95 patients. The primary endpoint was event-free survival (EFS), with secondary endpoints including 1-year EFS, 1-year overall survival (OS), disease control rate (DCR), and the correlation between MHC-II expression and clinico-biological characteristics. The cohort had a median age of 67 years (range, 33-90), with a male-to-female ratio of 50 : 45. Thirty-three percent of patients received the ipilimumab-nivolumab combination. The median follow-up was 16.8 months. Disease progression occurred in 58 patients (61%), with a median time to progression of 4.8 months. Forty-six patients (48.4%) experienced an event within the first year, and 52 patients (54.7%) died during follow-up. MHC-II positivity was observed in ≥10% of melanoma cells in 6.3% of patients. MHC-II expression was significantly associated with 1-year EFS ( P = 0.037) and DCR ( P = 0.032), but not with EFS or 1-year OS. Age, phototype, and brain metastases were correlated with MHC-II expression status. Our findings suggest that MHC-II expression by melanoma cells may serve as a favorable predictive biomarker for survival in metastatic cutaneous melanoma patients treated with ICIs.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Pierre Le Noac'h
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Pathology, University Hospital of Brest
| | - Patrice Hemon
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest
- Inserm, UMR1101, LaTIM, University of Western Brittany
| | - Laurent Misery
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Delphine Legoupil
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Christian Berthou
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| |
Collapse
|
2
|
van Dijk B, Janssen JC, van Daele PLA, de Jonge MJA, Joosse A, Verheul HMW, Epker JL, van der Veldt AAM. From ICI to ICU: A systematic review of patients with solid tumors who are treated with immune checkpoint inhibitors (ICI) and admitted to the intensive care unit (ICU). Cancer Treat Rev 2025; 136:102936. [PMID: 40222269 DOI: 10.1016/j.ctrv.2025.102936] [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: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have improved the survival of patients with different solid tumors and even resulted in cure of metastatic disease. Since the introduction of ICIs, an increasing number of patients is admitted to the ICU for severe and potentially life-threatening immune related adverse events (irAEs). The outcome of patients who are admitted to the ICU because of severe irAEs is still unknown. The aim of this systematic review is to collect evidence on the outcomes of patients with solid tumors who are admitted to the ICU because of irAEs. METHODS Medline, Embase, Cochrane central register of controlled trials and Google Scholar were searched systematically from 1975 to 24 September 2024. Articles were only included when describing patients with solid tumors who were admitted to the ICU because of irAEs after treatment with ICIs. Two independent reviewers extracted the data and assessed the risk of bias. RESULTS A total of 183 articles were included: two prospective ICU population-based studies, four retrospective ICU population-based studies, 25 retrospective studies describing irAEs with incidental ICU admissions, one review of case reports, and 153 articles with a total of 177 case reports. The six ICU population-based studies contained a total of 169 patients who were admitted to the ICU due to irAEs. In these six studies, the most frequently reported irAEs were pneumonitis and neurological irAEs. Of these 169 patients, 26% of the patients died on the ICU and an additional 8% of patients in the three to six months thereafter due to irAEs or disease progression. In all 183 included articles, various irAEs were described and the reported mortality rate varied from 0 to 53%. CONCLUSION The potential favorable outcomes of both the solid tumors and irAEs will probably result in more need for ICU admissions. Prospective clinical trials are needed to optimize the treatment strategy of severe irAEs at the ICU. Based on the favourable outcomes after life-threatening irAEs, ICU admission should definitely be considered for patients with solid tumors who have life-threatening irAEs.
Collapse
Affiliation(s)
- Brigit van Dijk
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Joséphine C Janssen
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Oncological Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Paul L A van Daele
- Department of Clinical Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Jelle L Epker
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Lavender V, Tanay M, Gomm L. The Transformation of Cancer Nursing Propelled by Novel SACT. Semin Oncol Nurs 2025; 41:151843. [PMID: 39952818 DOI: 10.1016/j.soncn.2025.151843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Verna Lavender
- Head of Nursing for Research and Education, Cancer and Surgery, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Mary Tanay
- Nurse Consultant (Genomics), Berkshire Cancer Centre, Royal Berkshire NHS Foundation Trust, London, UK
| | - Linda Gomm
- Advanced Nurse Practitioner (Cell Therapy & Early Phase Trials), Oncology and Haematology Clinical Trials, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
4
|
Zhang B. Overview of systemic anticancer treatments: conventional cytotoxics. Drug Ther Bull 2025; 63:37-43. [PMID: 39904574 DOI: 10.1136/dtb.2023.000059] [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: 02/06/2025]
Abstract
Cancer treatment is rapidly evolving and this review provides healthcare professionals who are not specialists in cancer therapeutics with a broad overview of the role of cancer systemic therapy, with a particular focus on chemotherapy.Historically, the majority of cytotoxic chemotherapy was used in patients with incurable or metastatic disease with the goal of disease control and symptom palliation. Now, with the advent of more effective, targeted systemic therapies (incorporating both cytotoxic and non-cytotoxic agents), systemic therapies are being used in more diverse treatment settings, both to increase the likelihood of cure and to induce prolonged disease remission.Chemotherapy (henceforth referring specifically to cytotoxic chemotherapy) remains important for the treatment of many cancer types. This article will review the principles of chemotherapy and the first-line systemic treatment paradigm of different cancer types. The potential toxicities of chemotherapy will also be described.
Collapse
Affiliation(s)
- Betty Zhang
- Department of Medical Oncology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
- Medical Oncology Research Fellow, Australian Rare Cancer Portal, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Centre for Molecular oncology, School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Placke JM, Rajcsanyi LS, Herbst R, Terheyden P, Utikal J, Pföhler C, Kreuter A, Mohr P, Gutzmer R, Weichenthal M, Meier F, Berking C, Leiter U, Seier J, Krefting F, Tasdogan A, Lodde GC, Livingstone E, Zimmer L, Roesch A, Griewank K, Schadendorf D, Ugurel S. Presence of brain metastasis differentially impacts long-term survival after first-line therapy in melanoma depending on BRAF mutation status. Front Immunol 2025; 16:1536642. [PMID: 40028330 PMCID: PMC11868123 DOI: 10.3389/fimmu.2025.1536642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Background Modern therapeutic strategies have significantly improved the prognosis of advanced melanoma patients. Predictive factors of therapy response include serum LDH; however, predictive markers for long-term survival are currently largely lacking. Patients and methods Patients diagnosed with stage IV melanoma (AJCCv8) of cutaneous origin or unknown primary were identified from the prospective multicenter German Dermatologic Cooperative Oncology Group (DeCOG) skin cancer registry ADOREG. Baseline characteristics were compared between patient groups with short-term versus long-term survival. Statistical analysis included ROC analysis and multinomial regression analysis. Results Of 3066 stage IV melanoma patients entered into the ADOREG between 05/2014 and 06/2021, 395 were identified for this study, of whom 301 (76.2%) survived ≤1 year, and 94 (23.8%) survived ≥5 years after stage IV diagnosis. The median follow-up time was 6 months (range 0-129 months). Regarding the baseline characteristics, only elevated serum LDH (P <0.001) was found to be independently predicting survival ≤1 year. Type of first-line therapy, immune checkpoint inhibition (ICI) versus BRAF/MEK targeted therapy (TT), was not predictive of long-term survival ≥5 years. For survival ≤1 year, the presence of brain metastases at treatment start was an independent predictor in BRAF-mutated patients regardless if they received TT (N=113; P=0<0.001) or ICI (N=69; P=0.015), but not in BRAF-wildtype patients who received ICI (N=161; P=0.47). Conclusions Low serum LDH independently predicts long-term survival of stage IV melanoma patients in every subgroup of treatment type and BRAF status. Brain metastasis has a negative impact on long-term survival in BRAF-mutated, but not in BRAF-wildtype patients. Investigation of molecular features of brain metastases in BRAF-mutated vs. BRAF-wildtype melanomas may lead to new insights in tumor biology and may yield new therapeutic approaches.
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Luisa Sophie Rajcsanyi
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Institute for Sex- and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| | - Rudolf Herbst
- Department of Dermatology, Helios Klinikum Erfurt, Erfurt, Germany
| | | | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/Saar, Germany
| | - Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Minden, Minden, Germany
| | - Michael Weichenthal
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, CCC Erlangen – EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Johanna Seier
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Frederik Krefting
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Georg C. Lodde
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
6
|
Zhou P, Wu Y, Han G, Jiang J, Wang H, Lu C, Liu Y, Wu J, Zou P, Wu H. Comparison of a Series of 68Ga-Labeled DOTA-LLP2A Conjugates for Positron Emission Tomography Imaging of Very Late Antigen-4 in Melanoma. Mol Pharm 2025; 22:1031-1041. [PMID: 39748758 DOI: 10.1021/acs.molpharmaceut.4c01204] [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: 01/04/2025]
Abstract
Melanoma, with its steadily rising global incidence, is characterized by high invasiveness, leading to poor prognosis in advanced stages. There remains an unmet clinical need for the development of radiolabeled PET imaging probes for the early diagnosis of melanoma. Integrin VLA-4, a key factor in melanoma metastasis, presents a promising protein target to address the specificity shortcomings of existing probes in melanoma imaging. This study evaluates 68Ga-labeled DOTA-LLP2A PET probes for melanoma imaging by modifying different carboxyl sites and employing various polyethylene glycol (PEG) linkers based on the structure of the high-affinity ligand LLP2A for VLA-4. The ligand intermediates LLP2A-NH2 and LLP2A(tBu)-OH, as well as their conjugates (probe precursors), were synthesized via solid-phase synthesis. The specificity and cytotoxicity of the probes were assessed in VLA-4-positive B16F10 cells and VLA-4-negative A375 cells. Targeting efficacy of the probes in B16F10 and A375 xenograft models was compared through PET imaging and biodistribution studies. VLA-4 expression in tissues was evaluated via immunofluorescence, while H&E staining was employed to assess the safety profile of the probes. The probe ([68Ga]Ga-T-CH) modified at the Aminocyclohexane carboxylic acid (Ach) exhibited greater signal accumulation in B16F10 melanoma (3.90 ± 0.43%ID/g at 1 h) compared to the 2-aminoadipic acid (Aad) side-chain-modified probe ([68Ga]Ga-T-AD) (1.43 ± 0.23%ID/g at 1 h). PET images of the three PEG conjugates derived from the Ach demonstrated bright tumor signals and low background noise, showing a progressive increase in tumor signal intensity from [68Ga]Ga-T6 to [68Ga]Ga-T4 and [68Ga]Ga-T2. Tumor uptake, tumor-to-muscle ratio, and tumor-to-blood ratio from biodistribution were significantly higher for [68Ga]Ga-T2 than for [68Ga]Ga-T4 and [68Ga]Ga-T6 (tumor: 3.58 ± 0.28 vs 2.90 ± 0.16 vs 1.87 ± 0.22%ID/g at 1 h; tumor/muscle: 13.38 ± 0.43 vs 10.62 ± 0.70 vs 7.19 ± 1.15 at 1 h; tumor/blood: 8.64 ± 1.12 vs 5.32 ± 0.91 vs 4.36 ± 0.59 at 1 h; P < 0.05). These data suggest that the series of PEG derivatives [68Ga]Ga-T2, [68Ga]Ga-T4, and [68Ga]Ga-T6, linked at the Ach site, are excellent 68Ga-labeled probes for melanoma and other potential VLA-4-positive tumors. Among them, [68Ga]Ga-T2 shows the highest tumor-to-background contrast for melanoma, positioning it as the most promising candidate for clinical translation.
Collapse
Affiliation(s)
- Peng Zhou
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yujing Wu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Guoqing Han
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Juntao Jiang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hongyong Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Chunxiong Lu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yaling Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Jun Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Pei Zou
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Hao Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
7
|
Sherapura A, Kiran BK, Pavan Kumar GS, Siddesh BM, Thirusangu P, Suchetha Kumari N, Prabhakar BT. Withaferin-A induced vimentin S56 phosphorylation dissociates NEDD9 signaling loop to regress progressive metastatic melanoma into lung adenocarcinoma. Chem Biol Interact 2025; 406:111319. [PMID: 39613173 DOI: 10.1016/j.cbi.2024.111319] [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: 09/15/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Metastasis is complex and insidious type of disease involves multiple signaling nexus, which have implications in understanding disease pathogenesis. Treatment failure for metastatic cancer is frequently high due to aggressive adaptation of cancerous cells to invade to neighboring organs. Cytoskeleton intermediate filamentous protein Vimentin and scaffolding protein Neural precursor cell expressed Developmentally Down-regulated protein 9 (NEDD9) play a key role in metastatic events by regulating multiple metastatic events. Interaction between these proteins is necessary to promote metastatic progression. Withaferin A (WFA), a natural pharamacophore, known to target Vimentin to induce antitumor potential. However exact molecular mechanism still yet to be elucidated. We hypothesize, Vimentin-NEDD9 signaling nexus is necessary for metastatic progression and targeting this interwoven signaling loop with effective pharamacophore WFA halts metastatic progression of melanoma into lung. To elucidate the same, we carried out gene expression measurement through quantitative Reverses Transcription Polymerase Chain Reaction (qRT-PCR), Immunoblot and Immunohistochemistry. Assessment of interactive signaling by Co-immunoprecipitation, Immunofluorescence, Co-localization and Proximity ligation assay. Phosphorylation studies through transfection of phospho specific mutant constructs generated through site directed mutagenesis. WFA induced cellular behavioral changes by migration, invasion assays and Immunoblot analysis. The B16F10 induced mouse metastatic melanoma model to asses NEDD9-Vimentin expression and anti-metastasis induced by WFA. The results postulates, elevated levels and interaction between NEDD9-Vimentin proteins, have positive correlation in metastatic progression of melanoma into lung in both in-vitro and in-vivo condition, establishing it as therapeutic target. Pharmacologically, WFA targets this complex by extending its activity by not only inducing specific Serine 56 phosphorylation of Vimentin, also dissociates NEDD9 signaling loop to halt Epithelial-mesenchymal transition (EMT) and subsequent metastatic events. Eventually, modulation of the relevant metastatic genes E-Cadherin, N-Cadherin, SNAIL, MMP-2 & MMP-9 resulted in regression of metastatic melanoma progression to lung. The study validates WFA induced S56 phosphorylation is necessary to abrupt the NEDD9-Vimentin metastatic signaling complex to regress aggressive metastatic melanoma. The investigation emphasized more mechanistic approach of WFA. Understanding and targeting such integrative mechanical input in the tumor microenvironment will be a better therapeutic strategy to combat metastasis.
Collapse
Affiliation(s)
- Ankith Sherapura
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - B K Kiran
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - G S Pavan Kumar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - B M Siddesh
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India
| | - Prabhu Thirusangu
- Department of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - N Suchetha Kumari
- Department of Biochemistry, K. S. Hegde Medical Academy, NITTE University, Mangalore, Karnataka, India
| | - B T Prabhakar
- Molecular Biomedicine Laboratory, Postgraduate Department of Studies and Research in Biotechnology, Sahyadri Science College, Kuvempu University, Shivamogga, 577203, Karnataka, India.
| |
Collapse
|
8
|
Kim J, Brunetti B, Kumar A, Mangla A, Honda K, Yoshida A. Inhibition of glutaminase elicits senolysis in therapy-induced senescent melanoma cells. Cell Death Dis 2024; 15:902. [PMID: 39695080 DOI: 10.1038/s41419-024-07284-3] [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: 07/09/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
The cyclin D1-Cyclin-Dependent Kinases 4 and 6 (CDK4/6) complex is crucial for the development of melanoma. We previously demonstrated that targeting CDK4/6 using small molecule inhibitors (CDK4/6i) suppresses BrafV600E melanoma growth in vitro and in vivo through induction of cellular senescence. However, clinical trials investigating CDK4/6i in melanoma have not yielded successful outcomes, underscoring the necessity to enhance the therapeutic efficacy of CDK4/6i. Accumulated research has shown that while senescence initially suppresses cell proliferation, a prolonged state of senescence eventually leads to tumor relapse by altering the tumor microenvironment, suggesting that removal of those senescent cells (in a process referred to as senolysis) is of clinical necessity to facilitate clinical response. We demonstrate that glutaminase 1 (GLS1) expression is specifically upregulated in CDK4/6i-induced senescent BrafV600E melanoma cells. Upregulated GLS1 expression renders BrafV600E melanoma senescent cells vulnerable to GLS1 inhibitor (GLS1i). Furthermore, we demonstrate that this senolytic approach targeting upregulated GLS1 expression is applicable even though those cells developed resistance to the BrafV600E inhibitor vemurafenib, a frequently encountered substantial clinical challenge to treating patients. Thus, this novel senolytic approach may revolutionize current CDK4/6i mediated melanoma treatment if melanoma cells undergo senescence prior to developing resistance to CDK4/6i. Given that we demonstrate that a low dose of vemurafenib induced senescence, which renders BrafV600E melanoma cells susceptible to GLS1i and recent accumulated research shows many cancer cells undergo senescence in response to chemotherapy, radiation, and immunotherapy, this senolytic therapy approach may prove applicable to a wide range of cancer types once senescence and GLS1 expression are induced.
Collapse
Affiliation(s)
- Justin Kim
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Bryce Brunetti
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ayanesh Kumar
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ankit Mangla
- Department of Hematology and Oncology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Kord Honda
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Akihiro Yoshida
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
9
|
Dirven I, Pierre E, Vander Mijnsbrugge AS, Vounckx M, Kessels JI, Neyns B. Regorafenib Combined with BRAF/MEK Inhibitors for the Treatment of Refractory Melanoma Brain Metastases. Cancers (Basel) 2024; 16:4083. [PMID: 39682270 DOI: 10.3390/cancers16234083] [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: 10/28/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND There are no active treatment options for patients with progressive melanoma brain metastases (MBM) failing immune checkpoint blockade (ICB) and BRAF/MEK inhibitors (BRAF/MEKi). Regorafenib (REGO), an oral multi-kinase inhibitor (incl. RAF-dimer inhibition), can overcome adaptive resistance to BRAF/MEKi in preclinical models. METHODS This is a single-center retrospective case series of patients with refractory MBM treated with REGO plus BRAF/MEKi (compassionate use). RESULTS A total of 22 patients were identified (18 BRAF-mutant, 4 NRASQ61-mutant; 19 with progressive MBM; 11 on corticosteroids). Thirteen BRAFV600-mutant patients were progressing on BRAF/MEKi at the time of REGO association. BRAF-mutant patients received REGO (40-80 mg once daily) combined with BRAF/MEKi, NRAS-mutant patients were treated with REGO + MEKi (+low-dose BRAFi to mitigate skin-toxicity). Grade 3 TRAE included arterial hypertension (n = 4) and maculopapular rash (n = 3). There were no G4/5 TRAE. In BRAF-mutant patients, overall and intracranial objective response rates (overall ORR and IC-ORR) were 11 and 29%, and overall and intracranial disease control rates (overall DCR and IC-DCR) were 44 and 59%, respectively. In NRAS-mutant patients overall ORR and IC-ORR were 0 and 25% and overall DCR and IC-DCR were 25 and 50%, respectively. The median PFS and OS were, respectively, 7.1 and 16.4 weeks in BRAF-mutant and 8.6 and 10.1 weeks in NRAS-mutant patients. CONCLUSIONS In heavily pretreated patients with refractory MBM, REGO combined with BRAF/MEKi demonstrated promising anti-tumor activity with an acceptable safety profile. In BRAFV600-mutant melanoma patients, responses cannot solely be attributed to BRAF/MEKi rechallenge. Further investigation in a prospective trial is ongoing to increase understanding of the efficacy.
Collapse
Affiliation(s)
- Iris Dirven
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Eden Pierre
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - An-Sofie Vander Mijnsbrugge
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Manon Vounckx
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Jolien I Kessels
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Bart Neyns
- Team Laboratory for Medical and Molecular Oncology (LMMO), Translational Oncology Research Center (TORC), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
10
|
Mu X, Zhou Y, Yu Y, Zhang M, Liu J. The roles of cancer stem cells and therapeutic implications in melanoma. Front Immunol 2024; 15:1486680. [PMID: 39611156 PMCID: PMC11602477 DOI: 10.3389/fimmu.2024.1486680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Melanoma is a highly malignant skin tumor characterized by high metastasis and poor prognosis. Recent studies have highlighted the pivotal role of melanoma stem cells (MSCs)-a subpopulation of cancer stem cells (CSCs)-in driving tumor growth, metastasis, therapeutic resistance, and recurrence. Similar to CSCs in other cancers, MSCs possess unique characteristics, including specific surface markers, dysregulated signaling pathways, and the ability to thrive within complex tumor microenvironment (TME). This review explored the current landscape of MSC research, discussing the identification of MSC-specific surface markers, the role of key signaling pathways such as Wnt/β-catenin, Notch, and Hedgehog (Hh), and how interactions within the TME, including hypoxia and immune cells, contribute to MSC-mediated drug resistance and metastatic behavior. Furthermore, we also investigated the latest therapeutic strategies targeting MSCs, such as small-molecule inhibitors, immune-based approaches, and novel vaccine developments, with an emphasis on their potential to overcome melanoma progression and improve clinical outcomes. This review aims to provide valuable insights into the complex roles of MSCs in melanoma biology and offers perspectives for future research and therapeutic advances against this challenging disease.
Collapse
Affiliation(s)
- Xiaoli Mu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Zhou
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongxin Yu
- The Department of Plastic and Reconstructive Surgery, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyi Zhang
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiyan Liu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Taylor K, Latimer NR, Douglas T, Hatswell AJ, Ho S, Okorogheye G, Borril J, Chen C, Kim I, Bertwistle D. Treatment Effect Waning in Immuno-oncology Health Technology Assessments: A Review of Assumptions and Supporting Evidence with Proposals to Guide Modelling. PHARMACOECONOMICS 2024; 42:1181-1196. [PMID: 39177877 PMCID: PMC11499331 DOI: 10.1007/s40273-024-01423-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/24/2024]
Abstract
Treatment effect waning (TEW) refers to the attenuation of treatment effects over time. Assumptions of a sustained immuno-oncologic treatment effect have been a source of contention in health technology assessment (HTA). We review how TEW has been addressed in HTA and in the wider scientific literature. We analysed company submissions to English language HTA agencies and summarised methods and assumptions used. We subsequently reviewed TEW-related work in the ISPOR Scientific Presentations Database and conducted a targeted literature review (TLR) for evidence of the maintenance of immuno-oncology (IO) treatment effects post-treatment discontinuation. We found no standardised approach adopted by companies in submissions to HTA agencies, with immediate TEW most used in scenario analyses. Independently fitted survival models do however suggest TEW may often be implicitly modelled. Materials in the ISPOR scientific database suggest gradual TEW is more plausible than immediate TEW. The TLR uncovered evidence of durable survival in patients treated with IOs but no evidence that directly addresses the presence or absence of TEW. Our HTA review shows the need for a consistent and appropriate implementation of TEW in oncology appraisals. However, the TLR highlights the absence of direct evidence on TEW in literature, as TEW is defined in terms of relative treatment effects-not absolute survival. We propose a sequence of steps for analysts to use when assessing whether a TEW scenario is necessary and appropriate to present in appraisals of IOs.
Collapse
Affiliation(s)
| | - Nicholas R Latimer
- Delta Hat Limited, Nottingham, UK
- Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | | | - Anthony J Hatswell
- Delta Hat Limited, Nottingham, UK
- Department of Statistical Science, University College London, London, UK
| | - Sophia Ho
- Bristol Myers Squibb, Uxbridge, London, UK
| | | | | | - Clara Chen
- Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Inkyu Kim
- Bristol Myers Squibb, Lawrenceville, NJ, USA
| | | |
Collapse
|
12
|
Tutic-Sorrentino L, Cazzaniga S, Feldmeyer L, Benzaquen M. Positron emission tomography-computed tomography vs. brain magnetic resonance imaging for the detection of cerebral metastases of melanoma: a 5-year retrospective study. Clin Exp Dermatol 2024; 49:1179-1185. [PMID: 38624009 DOI: 10.1093/ced/llae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Patients with melanoma present a high risk of developing extracutaneous metastases. Positron emission tomography--computed tomography (PET-CT) is one of the preferred examinations for the staging of oncological patients. It is not the method of choice to detect brain metastases, but this technique has shown significant improvement and allows the detection of some of them. However, it is unclear how it performs compared with magnetic resonance imaging (MRI), the current gold standard for diagnosing brain metastases. OBJECTIVES To compare the accuracy of PET-CT and cerebral MRI to detect brain metastases in patients with melanoma. METHODS We retrospectively included all patients diagnosed with melanoma stage IIC-IV (American Joint Committee on Cancer 8th Edition, 2017) who presented at the skin tumour board of the University Hospital of Bern between January 2018 and December 2022. All radiological reports extracted from the patient management system were analysed to assess discrepancy between the visibility of brain metastases on PET-CT and brain MRI. RESULTS In this study including 393 patients, brain MRI demonstrated significantly better performance than PET-CT in detecting brain metastases. In 47 patients, cerebral metastases were detected completely, detected partially, or not detected by PET-CT in 2 (4%), 15 (32%) and 30 (64%), respectively. CONCLUSIONS Despite the increasing performance of PET-CT, this study highlights the crucial role of brain MRI, which remains the gold standard to detect cerebral metastases. Brain MRI should be performed in patients with high-risk melanoma from stage IIC to exclude brain metastases.
Collapse
Affiliation(s)
- Lisa Tutic-Sorrentino
- Department of Dermatology, Inselspital - Bern University Hospital, University of Bern, Bern, Switzerland
| | - Simone Cazzaniga
- Department of Dermatology, Inselspital - Bern University Hospital, University of Bern, Bern, Switzerland
- Centro Studi GISED, Bergamo, Italy
| | - Laurence Feldmeyer
- Department of Dermatology, Inselspital - Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michael Benzaquen
- Department of Dermatology, Inselspital - Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
de Groot TM, Sommerkamp AA, Thio QCBS, Karhade AV, Groot OQ, Oosterhof JHF, Ijpma FFA, VAN Ooijen PMA, Ploegmakers JJW, Jutte PC, Schwab JH, Doornberg JN. External validation of the SORG machine learning for 90-day and 1-year mortality in patients suffering from extremity metastatic disease in an European cohort of 174 patients. Acta Orthop Belg 2024; 90:493-501. [PMID: 39851022 DOI: 10.52628/90.3.12636] [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: 01/25/2025]
Abstract
Accurate survival prediction of patients with long-bone metastases is challenging, but important for optimizing treatment. The Skeletal Oncology Research Group (SORG) machine learning algorithm (MLA) has been previously developed and internally validated to predict 90-day and 1-year survival. External validation showed promise in the United States and Taiwan. To ensure global generalizability, the algorithm remains to be validated in Europe. We therefore asked: does the SORG-MLA for long-bone metastases accurately predict 90-day and 1-year survival in a European cohort? One-hundred seventy-four patients undergoing surgery for long-bone metastases between 1997-2019 were included at a tertiary referral Orthopaedic Oncology Center in the Netherlands. Model performance measures included discrimination, calibration, overall performance, and decision curve analysis. The SORG-MLA retained reasonable discriminative ability, showing an area under the curve of 0.73 for 90-day mortality and 0.77 for 1-year mortality. However, the calibration analysis demonstrated overestimation of European patients' 90- day mortality (calibration intercept -0.54, slope 0.60). For 1-year mortality (calibration intercept 0.01, slope 0.60) this was not the case. The Brier score predictions were lower than their respective null model (0.13 versus 0.14 for 90-day; 0.20 versus 0.25 for 1-year), suggesting good overall performance of the SORG-MLA for both timepoints. The SORG-MLA showed promise in predicting survival of patients with extremity metastatic disease. However, clinicians should keep in mind that due to differences in patient population, the model tends to underestimate survival in this Dutch cohort. The SORG model can be accessed freely at https://sorg-apps.shinyapps.io/extremitymetssurvival/.
Collapse
|
14
|
Amrane K, Meur CL, Thuillier P, Berthou C, Uguen A, Deandreis D, Bourhis D, Bourbonne V, Abgral R. Review on radiomic analysis in 18F-fluorodeoxyglucose positron emission tomography for prediction of melanoma outcomes. Cancer Imaging 2024; 24:87. [PMID: 38970050 PMCID: PMC11225300 DOI: 10.1186/s40644-024-00732-5] [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: 11/02/2023] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
Over the past decade, several strategies have revolutionized the clinical management of patients with cutaneous melanoma (CM), including immunotherapy and targeted tyrosine kinase inhibitor (TKI)-based therapies. Indeed, immune checkpoint inhibitors (ICIs), alone or in combination, represent the standard of care for patients with advanced disease without an actionable mutation. Notably BRAF combined with MEK inhibitors represent the therapeutic standard for disease disclosing BRAF mutation. At the same time, FDG PET/CT has become part of the routine staging and evaluation of patients with cutaneous melanoma. There is growing interest in using FDG PET/CT measurements to predict response to ICI therapy and/or target therapy. While semiquantitative values such as standardized uptake value (SUV) are limited for predicting outcome, new measures including tumor metabolic volume, total lesion glycolysis and radiomics seem promising as potential imaging biomarkers for nuclear medicine. The aim of this review, prepared by an interdisciplinary group of experts, is to take stock of the current literature on radiomics approaches that could improve outcomes in CM.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix, 29600, France.
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France.
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest, Brest, France
| | - Philippe Thuillier
- Department of Endocrinology, University Hospital of Brest, Brest, France
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
| | - Christian Berthou
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Lymphocytes B et Autoimmunité, Inserm, UMR1227, Univ Brest, Inserm, LabEx IGO, Brest, France
- Department of Pathology, University Hospital of Brest, Brest, France
| | - Désirée Deandreis
- Department of Nuclear Medicine, Gustave Roussy Institute, University of Paris Saclay, Paris, France
| | - David Bourhis
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| | - Vincent Bourbonne
- Department of Radiotherapy, University Hospital of Brest, Brest, France
- Inserm, UMR1101, LaTIM, University of Western Brittany, Brest, France
| | - Ronan Abgral
- UMR Inserm 1304 GETBO, University of Western Brittany, Brest, IFR 148, France
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
| |
Collapse
|
15
|
Lins FV, Bispo ECI, Rodrigues NS, Silva MVS, Carvalho JL, Gelfuso GM, Saldanha-Araujo F. Ibrutinib Modulates Proliferation, Migration, Mitochondrial Homeostasis, and Apoptosis in Melanoma Cells. Biomedicines 2024; 12:1012. [PMID: 38790974 PMCID: PMC11117653 DOI: 10.3390/biomedicines12051012] [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: 04/07/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Ibrutinib, a tyrosine kinase inhibitor with a broad spectrum of action, has been successfully explored to treat hematological and solid cancers. Herein, we investigated the anti-cancer effect of Ibrutinib on melanoma cell lines. Cytotoxicity was evaluated using the MTT assay. Apoptosis, mitochondrial membrane potential, reactive oxygen species (ROS) production, cell proliferation, and cell cycle stages were determined by flow cytometry. LDH release and Caspase 3/7 activity were determined by colorimetric and luminescent assays, respectively. Cell migration was evaluated by wound scratch assay. Gene expression was determined by real-time PCR. Gene Ontology (GO) enrichment analysis of melanoma clinical samples was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). MTT assays showed that Ibrutinib is toxic for MeWo, SK-MEL-28, and WM164 cells. The annexin V/PI staining, Caspase 3/7 activity, and LDH release in MeWo cells revealed that apoptosis is the primary mechanism of death caused by Ibrutinib. Corroborating such observation, we identified that Ibrutinib treatment impairs the mitochondrial membrane potential of such cells and significantly increases the transcriptional levels of the pro-apoptotic factors ATM, HRK, BAX, BAK, CASP3, and CASP8. Furthermore, Ibrutinib showed antimetastatic potential by inhibiting the migration of MeWo cells. Finally, we performed a functional enrichment analysis and identified that the differential expression of Ibrutinib-target molecules is associated with enrichment of apoptosis and necrosis pathways in melanoma samples. Taken together, our results clearly suggest that Ibrutinib can be successfully explored as an effective therapeutic approach for melanomas.
Collapse
Affiliation(s)
- Fernanda Vitelli Lins
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabete Cristina Iseke Bispo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Naomí Souza Rodrigues
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Maria Victória Souto Silva
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Juliana Lott Carvalho
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Guilherme Martins Gelfuso
- Laboratório de Medicamentos, Alimentos e Cosméticos, Universidade de Brasília, Brasília 70910-900, DF, Brazil;
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| |
Collapse
|
16
|
Wu Y, Yin S, Li C, Zhao L, Song M, Yu Y, Tang L, Yang Y. A signature of seven hypoxia-related lncRNAs is a potential biomarker for predicting the prognosis of melanoma. Am J Cancer Res 2024; 14:1712-1729. [PMID: 38726277 PMCID: PMC11076246 DOI: 10.62347/lhkw3124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Melanoma is the most aggressive type of skin cancer and has a high mortality rate once metastasis occurs. Hypoxia is a universal characteristic of the microenvironment of cancer and a driver of melanoma progression. In recent years, long noncoding RNAs (lncRNAs) have attracted widespread attention in oncology research. In this study, screening was performed and revealed seven hypoxia-related lncRNAs AC008687.3, AC009495.1, AC245128.3, AL512363.1, LINC00518, LINC02416 and MCCC1-AS1 as predictive biomarkers. A predictive risk model was constructed via univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses. Patients were grouped according to the model risk score, and Kaplan-Meier analysis was performed to compare survival between groups. Functional and pathway enrichment analyses were performed to compare gene set enrichment between groups. Moreover, a nomogram was constructed with the risk score as a variable. In both the training and validation sets, patients in the low-risk group had better overall survival than did those in the high-risk group (P<0.001). The 3-, 5- and 10-year area under the curve (AUC) values for the nomogram model were 0.821, 0.795 and 0.820, respectively. Analyses of immune checkpoints, immunotherapy response, drug sensitivity, and mutation landscape were also performed. The results suggested that the low-risk group had a better response to immunotherapeutic. In addition, the nomogram can effectively predict the prognosis and immunotherapy response of melanoma patients. The signature of seven hypoxia-related lncRNAs showed great potential value as an immunotherapy response biomarker, and these lncRNAs might be treatment targets for melanoma patients.
Collapse
Affiliation(s)
- Yunyang Wu
- School of Traditional Chinese Medicine, Naval Medical UniversityShanghai, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical UniversityShanghai, China
| | - Shenhui Yin
- National Key Laboratory of Immunity & Inflammation, Naval Medical UniversityShanghai, China
| | - Chunzhen Li
- National Key Laboratory of Immunity & Inflammation, Naval Medical UniversityShanghai, China
| | - Liyuan Zhao
- National Key Laboratory of Immunity & Inflammation, Naval Medical UniversityShanghai, China
| | - Mengqi Song
- National Key Laboratory of Immunity & Inflammation, Naval Medical UniversityShanghai, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical UniversityShanghai, China
| | - Ling Tang
- School of Traditional Chinese Medicine, Naval Medical UniversityShanghai, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical UniversityShanghai, China
| | - Yanlong Yang
- School of Traditional Chinese Medicine, Naval Medical UniversityShanghai, China
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical UniversityShanghai, China
| |
Collapse
|
17
|
Tittarelli A, Pereda C, Gleisner MA, López MN, Flores I, Tempio F, Lladser A, Achour A, González FE, Durán-Aniotz C, Miranda JP, Larrondo M, Salazar-Onfray F. Long-Term Survival and Immune Response Dynamics in Melanoma Patients Undergoing TAPCells-Based Vaccination Therapy. Vaccines (Basel) 2024; 12:357. [PMID: 38675738 PMCID: PMC11053591 DOI: 10.3390/vaccines12040357] [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: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer vaccines present a promising avenue for treating immune checkpoint blockers (ICBs)-refractory patients, fostering immune responses to modulate the tumor microenvironment. We revisit a phase I/II trial using Tumor Antigen-Presenting Cells (TAPCells) (NCT06152367), an autologous antigen-presenting cell vaccine loaded with heat-shocked allogeneic melanoma cell lysates. Initial findings showcased TAPCells inducing lysate-specific delayed-type hypersensitivity (DTH) reactions, correlating with prolonged survival. Here, we extend our analysis over 15 years, categorizing patients into short-term (<36 months) and long-term (≥36 months) survivors, exploring novel associations between clinical outcomes and demographic, genetic, and immunologic parameters. Notably, DTHpos patients exhibit a 53.1% three-year survival compared to 16.1% in DTHneg patients. Extended remissions are observed in long-term survivors, particularly DTHpos/M1cneg patients. Younger age, stage III disease, and moderate immune events also benefit short-term survivors. Immunomarkers like increased C-type lectin domain family 2 member D on CD4+ T cells and elevated interleukin-17A were detected in long-term survivors. In contrast, toll-like receptor-4 D229G polymorphism and reduced CD32 on B cells are associated with reduced survival. TAPCells achieved stable long remissions in 35.2% of patients, especially M1cneg/DTHpos cases. Conclusions: Our study underscores the potential of vaccine-induced immune responses in melanoma, emphasizing the identification of emerging biological markers and clinical parameters for predicting long-term remission.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile;
| | - Cristian Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - María A. Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Mercedes N. López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Fabián Tempio
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580702, Chile;
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 8580702, Chile
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Fermín E. González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile;
| | - Claudia Durán-Aniotz
- Latin American Brain Health Institute (BrainLat), Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibañez, Santiago 7941169, Chile;
| | | | - Milton Larrondo
- Banco de Sangre, Hospital Clínico de la Universidad de Chile, Santiago 8380453, Chile;
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
18
|
Lim SY, Rizos H. Single-cell RNA sequencing in melanoma: what have we learned so far? EBioMedicine 2024; 100:104969. [PMID: 38241976 PMCID: PMC10831183 DOI: 10.1016/j.ebiom.2024.104969] [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: 10/29/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
Over the past decade, there have been remarkable improvements in the treatment and survival rates of melanoma patients. Treatment resistance remains a persistent challenge, however, and is partly attributable to intratumoural heterogeneity. Melanoma cells can transition through a series of phenotypic and transcriptional cell states that vary in invasiveness and treatment responsiveness. The diverse stromal and immune contexture of the tumour microenvironment also contributes to intratumoural heterogeneity and disparities in treatment response in melanoma patients. Recent advances in single-cell sequencing technologies have enabled a more detailed understanding of melanoma heterogeneity and the underlying transcriptional programs that regulate melanoma cell diversity and behaviour. In this review, we examine the concept of intratumoural heterogeneity and the challenges it poses to achieving long-lasting treatment responses. We focus on the significance of next generation single-cell sequencing in advancing our understanding of melanoma diversity and the unique insights gained from single-cell studies.
Collapse
Affiliation(s)
- Su Yin Lim
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia.
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia
| |
Collapse
|
19
|
Amrane K, Le Meur C, Besse B, Hemon P, Le Noac’h P, Pradier O, Berthou C, Abgral R, Uguen A. HLA-DR expression in melanoma: from misleading therapeutic target to potential immunotherapy biomarker. Front Immunol 2024; 14:1285895. [PMID: 38299143 PMCID: PMC10827890 DOI: 10.3389/fimmu.2023.1285895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Since the advent of anti-PD1 immune checkpoint inhibitor (ICI) immunotherapy, cutaneous melanoma has undergone a true revolution with prolonged survival, as available 5-year updates for progression-free survival and overall survival demonstrate a durable clinical benefit for melanoma patients receiving ICI. However, almost half of patients fail to respond to treatment, or relapse sooner or later after the initial response to therapy. Little is known about the reasons for these failures. The identification of biomarkers seems necessary to better understand this resistance. Among these biomarkers, HLA-DR, a component of MHC II and abnormally expressed in certain tumor types including melanoma for unknown reasons, seems to be an interesting marker. The aim of this review, prepared by an interdisciplinary group of experts, is to take stock of the current literature on the potential interest of HLA-DR expression in melanoma as a predictive biomarker of ICI outcome.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix, France
- Inserm, Unité mixte de recherche (UMR1227), Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx Immunotherapy-Graft-Oncology (IGO), Brest, France
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest, Brest, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy Cancer Centre, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Patrice Hemon
- Inserm, Unité mixte de recherche (UMR1227), Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx Immunotherapy-Graft-Oncology (IGO), Brest, France
| | - Pierre Le Noac’h
- Inserm, Unité mixte de recherche (UMR1227), Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx Immunotherapy-Graft-Oncology (IGO), Brest, France
- Department of Pathology, University Hospital of Brest, Brest, France
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest, Brest, France
| | - Christian Berthou
- Inserm, Unité mixte de recherche (UMR1227), Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx Immunotherapy-Graft-Oncology (IGO), Brest, France
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest, Brest, France
- UMR Inserm 1304 Groupe d'Étude de la Thrombose de Bretagne Occidentale (GETBO), IFR 148, University of Western Brittany, Brest, France
| | - Arnaud Uguen
- Inserm, Unité mixte de recherche (UMR1227), Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx Immunotherapy-Graft-Oncology (IGO), Brest, France
- Department of Pathology, University Hospital of Brest, Brest, France
| |
Collapse
|
20
|
Moreno-Ramírez D, Fernández-Orland A, Ferrándiz L. Immunotherapy and Targeted Therapy in Patients With Advanced Melanoma and the V600 BRAF Mutation: Which One First? ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:48-55. [PMID: 37321549 DOI: 10.1016/j.ad.2023.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.
Collapse
Affiliation(s)
- D Moreno-Ramírez
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España.
| | - A Fernández-Orland
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España
| | - L Ferrándiz
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Sevilla, España
| |
Collapse
|
21
|
Moreno-Ramírez D, Fernández-Orland A, Ferrándiz L. [Translated article] Immunotherapy and Targeted Therapy in Patients With Advanced Melanoma and the V600 BRAF Mutation: Which One First? ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T48-T55. [PMID: 37923078 DOI: 10.1016/j.ad.2023.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 11/07/2023] Open
Abstract
Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.
Collapse
Affiliation(s)
- D Moreno-Ramírez
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain.
| | - A Fernández-Orland
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - L Ferrándiz
- Unidad de Melanoma, Servicio de Dermatología Médico-Quirúrgica y Venereología, Hospital Universitario Virgen Macarena, Seville, Spain
| |
Collapse
|
22
|
Dayimu A, Gupta A, Matin RN, Nobes J, Board R, Payne M, Rao A, Fusi A, Danson S, Eccles B, Carser J, Brown CO, Steven N, Bhattacharyya M, Brown E, Gonzalez M, Highley M, Pickering L, Kumar S, Waterston A, Burghel G, Demain L, Baker E, Wulff J, Qian W, Twelves S, Middleton M, Corrie P. A randomised phase 2 study of intermittent versus continuous dosing of dabrafenib plus trametinib in patients with BRAF V600 mutant advanced melanoma (INTERIM). Eur J Cancer 2024; 196:113455. [PMID: 38029480 DOI: 10.1016/j.ejca.2023.113455] [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: 09/05/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND BRAF+MEK inhibitors extend life expectancy of patients with BRAFV600 mutant advanced melanoma. Acquired resistance limits duration of benefit, but preclinical and case studies suggest intermittent dosing could overcome this limitation. INTERIM was a phase 2 trial evaluating an intermittent dosing regimen. METHODS Patients with BRAFV600 mutant advanced melanoma due to start dabrafenib+trametinib were randomised to receive either continuous (CONT), or intermittent (INT; dabrafenib d1-21, trametinib d1-14 every 28 days) dosing. A composite primary endpoint included progression-free survival (PFS) and quality of life (QoL). Secondary endpoints included response rate (ORR), overall survival (OS) and adverse events (AEs). Mutant BRAFV600E ctDNA was measured by droplet digital PCR (ddPCR), using mutant allele frequency of > 1 % as the detection threshold. RESULTS 79 patients (39 INT, 40 CONT) were recruited; median age 67 years, 65 % AJCC (7th ed) stage IV M1c, 29 % had brain metastases. With 19 months median follow-up, INT was inferior in all efficacy measures: median PFS 8.5 vs 10.7mo (HR 1.39, 95 %CI 0.79-2.45, p = 0.255); median OS 18.1mo vs not reached (HR 1.69, 95 %CI 0.87-3.28, p = 0.121), ORR 57 % vs 77 %. INT patients experienced fewer treatment-related AEs (76 % vs 88 %), but more grade > 3 AEs (53 % vs 42 %). QoL favoured CONT. Detection of BRAFV600E ctDNA prior to treatment correlated with worse OS (HR 2.55, 95 %CI 1.25-5.21, p = 0.01) in both arms. A change to undetected during treatment did not significantly predict better OS. CONCLUSION INTERIM findings are consistent with other recent clinical trials reporting that intermittent dosing does not improve efficacy of BRAF+MEK inhibitors.
Collapse
Affiliation(s)
- Alimu Dayimu
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Rubeta N Matin
- Department of Dermatology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jenny Nobes
- Department of Oncology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norfolk, UK
| | - Ruth Board
- Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Preston, UK
| | - Miranda Payne
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Ankit Rao
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Alberto Fusi
- Department of Medical Oncology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Sarah Danson
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK; Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Bryony Eccles
- Department of Medical Oncology, University Hospitals Dorset NHS Foundation Trust, Poole, UK
| | - Judith Carser
- Department of Oncology, Belfast Health and Social Care Trust, Belfast, UK
| | | | - Neil Steven
- Department of Oncology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Ewan Brown
- Western General Hospital, Lothian NHS Board, Edinburgh, UK
| | - Michael Gonzalez
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Martin Highley
- Oncology Centre, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Lisa Pickering
- Skin and Renal Units, Royal Marsden NHS Foundation Trust, London, UK
| | - Satish Kumar
- Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UK
| | | | - George Burghel
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Leigh Demain
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Eleanor Baker
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jerome Wulff
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Wendi Qian
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sophie Twelves
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mark Middleton
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK; Department of Oncology, University of Oxford, Oxford, UK
| | - Pippa Corrie
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
23
|
Shalata W, Abu Jama A, Abu Salman A, Golosky M, Solomon A, Abu Saleh O, Michlin R, Shalata S, Agbarya A, Yakobson A. Unexpected and Rare Sites of Metastasis in Oncologic Patients. J Clin Med 2023; 12:6447. [PMID: 37892585 PMCID: PMC10607747 DOI: 10.3390/jcm12206447] [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: 08/25/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Case studies of rare oncologic metastases are an important source of clinical data for health care professionals and researchers. While infrequent, the knowledge base and clinical recommendations derived from such cases aid in advancements in the field. As such, we aim to add five cases to the growing body of literature. The first two male patients, aged 69 and 73, were diagnosed with colon adenocarcinoma, suspected to be a second primary prostate carcinoma, following positron emission tomography-computer tomography (PET-CT). This suspicion was ruled out by prostatectomy and histopathological investigations, which instead found adenocarcinoma of colonic origin. The next two male patients, ages 63 and 68, were diagnosed, respectively, with metastatic pancreatic adenocarcinoma with cardiac metastases and metastatic melanoma with distant metastases to the pancreas. The final patient was a 73-year-old male diagnosed with metastatic breast cancer after a radiological investigation of suspected renal cell carcinoma.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (A.A.J.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (A.S.)
| | - Ashraf Abu Jama
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (A.A.J.)
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (A.S.)
| | - Amjad Abu Salman
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (A.S.)
- Cardiology Division, Soroka Medical Center, Beer Sheva 84105, Israel
| | - Mitchell Golosky
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (A.S.)
- Medical School for International Health and Sciences, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Adam Solomon
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel (A.S.)
- Medical School for International Health and Sciences, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Omar Abu Saleh
- Department of Dermatology and Venereology, The Emek Medical Centre, Afula 18341, Israel
| | - Regina Michlin
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (A.A.J.)
| | - Sondos Shalata
- Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel;
| | - Abed Agbarya
- Department of Oncology, Bnai Zion Medical Center, Haifa 31048, Israel
| | - Alexander Yakobson
- The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (A.A.J.)
| |
Collapse
|
24
|
Pavlick AC, Ariyan CE, Buchbinder EI, Davar D, Gibney GT, Hamid O, Hieken TJ, Izar B, Johnson DB, Kulkarni RP, Luke JJ, Mitchell TC, Mooradian MJ, Rubin KM, Salama AK, Shirai K, Taube JM, Tawbi HA, Tolley JK, Valdueza C, Weiss SA, Wong MK, Sullivan RJ. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of melanoma, version 3.0. J Immunother Cancer 2023; 11:e006947. [PMID: 37852736 PMCID: PMC10603365 DOI: 10.1136/jitc-2023-006947] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 10/20/2023] Open
Abstract
Since the first approval for immune checkpoint inhibitors (ICIs) for the treatment of cutaneous melanoma more than a decade ago, immunotherapy has completely transformed the treatment landscape of this chemotherapy-resistant disease. Combination regimens including ICIs directed against programmed cell death protein 1 (PD-1) with anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) agents or, more recently, anti-lymphocyte-activation gene 3 (LAG-3) agents, have gained regulatory approvals for the treatment of metastatic cutaneous melanoma, with long-term follow-up data suggesting the possibility of cure for some patients with advanced disease. In the resectable setting, adjuvant ICIs prolong recurrence-free survival, and neoadjuvant strategies are an active area of investigation. Other immunotherapy strategies, such as oncolytic virotherapy for injectable cutaneous melanoma and bispecific T-cell engager therapy for HLA-A*02:01 genotype-positive uveal melanoma, are also available to patients. Despite the remarkable efficacy of these regimens for many patients with cutaneous melanoma, traditional immunotherapy biomarkers (ie, programmed death-ligand 1 expression, tumor mutational burden, T-cell infiltrate and/or microsatellite stability) have failed to reliably predict response. Furthermore, ICIs are associated with unique toxicity profiles, particularly for the highly active combination of anti-PD-1 plus anti-CTLA-4 agents. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop this clinical practice guideline on immunotherapy for the treatment of melanoma, including rare subtypes of the disease (eg, uveal, mucosal), with the goal of improving patient care by providing guidance to the oncology community. Drawing from published data and clinical experience, the Expert Panel developed evidence- and consensus-based recommendations for healthcare professionals using immunotherapy to treat melanoma, with topics including therapy selection in the advanced and perioperative settings, intratumoral immunotherapy, when to use immunotherapy for patients with BRAFV600-mutated disease, management of patients with brain metastases, evaluation of treatment response, special patient populations, patient education, quality of life, and survivorship, among others.
Collapse
Affiliation(s)
| | - Charlotte E Ariyan
- Department of Surgery Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Diwakar Davar
- Hillman Cancer Center, University of Pittsburg Medical Center, Pittsburgh, Pennsylvania, USA
| | - Geoffrey T Gibney
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, California, USA
| | - Tina J Hieken
- Department of Surgery and Comprehensive Cancer Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rajan P Kulkarni
- Departments of Dermatology, Oncological Sciences, Biomedical Engineering, and Center for Cancer Early Detection Advanced Research, Knight Cancer Institute, OHSU, Portland, Oregon, USA
- Operative Care Division, VA Portland Health Care System (VAPORHCS), Portland, Oregon, USA
| | - Jason J Luke
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Tara C Mitchell
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Meghan J Mooradian
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Krista M Rubin
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - April Ks Salama
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, Carolina, USA
| | - Keisuke Shirai
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Janis M Taube
- Department of Dermatology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J Keith Tolley
- Patient Advocate, Melanoma Research Alliance, Washington, DC, USA
| | - Caressa Valdueza
- Cutaneous Oncology Program, Weill Cornell Medicine, New York, New York, USA
| | - Sarah A Weiss
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Michael K Wong
- Patient Advocate, Melanoma Research Alliance, Washington, DC, USA
| | - Ryan J Sullivan
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Castellani G, Buccarelli M, Arasi MB, Rossi S, Pisanu ME, Bellenghi M, Lintas C, Tabolacci C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers (Basel) 2023; 15:4026. [PMID: 37627054 PMCID: PMC10452867 DOI: 10.3390/cancers15164026] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer resulting from the malignant transformation of melanocytes. Recent therapeutic approaches, including targeted therapy and immunotherapy, have improved the prognosis and outcome of melanoma patients. BRAF is one of the most frequently mutated oncogenes recognised in melanoma. The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ERK (MAPK) signalling pathway. Therefore, mutated BRAF has become a useful target for molecular therapy and the use of BRAF kinase inhibitors has shown promising results. However, several resistance mechanisms invariably develop leading to therapeutic failure. The aim of this manuscript is to review the role of BRAF mutational status in the pathogenesis of melanoma and its impact on differentiation and inflammation. Moreover, this review focuses on the mechanisms responsible for resistance to targeted therapies in BRAF-mutated melanoma and provides an overview of circulating biomarkers including circulating tumour cells, circulating tumour DNA, and non-coding RNAs.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Beatrice Arasi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| | - Maria Elena Pisanu
- High Resolution NMR Unit, Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Bellenghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.C.); (M.B.); (M.B.A.); (S.R.)
| |
Collapse
|
26
|
Ascierto PA, Agarwala SS, Warner AB, Ernstoff MS, Fox BA, Gajewski TF, Galon J, Garbe C, Gastman BR, Gershenwald JE, Kalinski P, Krogsgaard M, Leidner RS, Lo RS, Menzies AM, Michielin O, Poulikakos PI, Weber JS, Caracò C, Osman I, Puzanov I, Thurin M. Perspectives in Melanoma: meeting report from the Melanoma Bridge (December 1st-3rd, 2022-Naples, Italy). J Transl Med 2023; 21:508. [PMID: 37507765 PMCID: PMC10375730 DOI: 10.1186/s12967-023-04325-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/30/2023] Open
Abstract
Outcomes for patients with melanoma have improved over the past decade with the clinical development and approval of immunotherapies targeting immune checkpoint receptors such as programmed death-1 (PD-1), programmed death ligand 1 (PD-L1) or cytotoxic T lymphocyte antigen-4 (CTLA-4). Combinations of these checkpoint therapies with other agents are now being explored to improve outcomes and enhance benefit-risk profiles of treatment. Alternative inhibitory receptors have been identified that may be targeted for anti-tumor immune therapy, such as lymphocyte-activation gene-3 (LAG-3), as have several potential target oncogenes for molecularly targeted therapy, such as tyrosine kinase inhibitors. Unfortunately, many patients still progress and acquire resistance to immunotherapy and molecularly targeted therapies. To bypass resistance, combination treatment with immunotherapies and single or multiple TKIs have been shown to improve prognosis compared to monotherapy. The number of new combinations treatment under development for melanoma provides options for the number of patients to achieve a therapeutic benefit. Many diagnostic and prognostic assays have begun to show clinical applicability providing additional tools to optimize and individualize treatments. However, the question on the optimal algorithm of first- and later-line therapies and the search for biomarkers to guide these decisions are still under investigation. This year, the Melanoma Bridge Congress (Dec 1st-3rd, 2022, Naples, Italy) addressed the latest advances in melanoma research, focusing on themes of paramount importance for melanoma prevention, diagnosis and treatment. This included sessions dedicated to systems biology on immunotherapy, immunogenicity and gene expression profiling, biomarkers, and combination treatment strategies.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | | | | | - Marc S Ernstoff
- ImmunoOncology Branch (IOB), Developmental Therapeutics Program, Cancer Therapy and Diagnosis Division, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Bernard A Fox
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Thomas F Gajewski
- Department of Pathology and Department of Medicine (Section of Hematology/Oncology), University of Chicago, Chicago, IL, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006, Paris, France
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Brian R Gastman
- Department of Surgery, School of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rom S Leidner
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Roger S Lo
- Jonsson Comprehensive Cancer Center David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Olivier Michielin
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Poulikos I Poulikakos
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, a NCI-Funded Comprehensive Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori "Fondazione Pascale" IRCCS, Naples, Italy
| | - Iman Osman
- Rudolf L, Baer, New York University Langone Medical Center, New York, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Magdalena Thurin
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
27
|
Yan C, Nebhan CA, Saleh N, Shattuck-Brandt R, Chen SC, Ayers GD, Weiss V, Richmond A, Vilgelm AE. Generation of Orthotopic Patient-Derived Xenografts in Humanized Mice for Evaluation of Emerging Targeted Therapies and Immunotherapy Combinations for Melanoma. Cancers (Basel) 2023; 15:3695. [PMID: 37509357 PMCID: PMC10377652 DOI: 10.3390/cancers15143695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Current methodologies for developing PDX in humanized mice in preclinical trials with immune-based therapies are limited by GVHD. Here, we compared two approaches for establishing PDX tumors in humanized mice: (1) PDX are first established in immune-deficient mice; or (2) PDX are initially established in humanized mice; then established PDX are transplanted to a larger cohort of humanized mice for preclinical trials. With the first approach, there was rapid wasting of PDX-bearing humanized mice with high levels of activated T cells in the circulation and organs, indicating immune-mediated toxicity. In contrast, with the second approach, toxicity was less of an issue and long-term human melanoma tumor growth and maintenance of human chimerism was achieved. Preclinical trials from the second approach revealed that rigosertib, but not anti-PD-1, increased CD8/CD4 T cell ratios in spleen and blood and inhibited PDX tumor growth. Resistance to anti-PD-1 was associated with PDX tumors established from tumors with limited CD8+ T cell content. Our findings suggest that it is essential to carefully manage immune editing by first establishing PDX tumors in humanized mice before expanding PDX tumors into a larger cohort of humanized mice to evaluate therapy response.
Collapse
Affiliation(s)
- Chi Yan
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Caroline A. Nebhan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
- Division of Hematology & Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nabil Saleh
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
| | - Rebecca Shattuck-Brandt
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.-C.C.); (G.D.A.)
| | - Gregory D. Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.-C.C.); (G.D.A.)
| | - Vivian Weiss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Ann Richmond
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; (C.Y.); (N.S.); (R.S.-B.)
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37232, USA;
| | - Anna E. Vilgelm
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center—Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH 43210, USA
| |
Collapse
|
28
|
Anderson KG, Braun DA, Buqué A, Gitto SB, Guerriero JL, Horton B, Keenan BP, Kim TS, Overacre-Delgoffe A, Ruella M, Triplett TA, Veeranki O, Verma V, Zhang F. Leveraging immune resistance archetypes in solid cancer to inform next-generation anticancer therapies. J Immunother Cancer 2023; 11:e006533. [PMID: 37399356 PMCID: PMC10314654 DOI: 10.1136/jitc-2022-006533] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 07/05/2023] Open
Abstract
Anticancer immunotherapies, such as immune checkpoint inhibitors, bispecific antibodies, and chimeric antigen receptor T cells, have improved outcomes for patients with a variety of malignancies. However, most patients either do not initially respond or do not exhibit durable responses due to primary or adaptive/acquired immune resistance mechanisms of the tumor microenvironment. These suppressive programs are myriad, different between patients with ostensibly the same cancer type, and can harness multiple cell types to reinforce their stability. Consequently, the overall benefit of monotherapies remains limited. Cutting-edge technologies now allow for extensive tumor profiling, which can be used to define tumor cell intrinsic and extrinsic pathways of primary and/or acquired immune resistance, herein referred to as features or feature sets of immune resistance to current therapies. We propose that cancers can be characterized by immune resistance archetypes, comprised of five feature sets encompassing known immune resistance mechanisms. Archetypes of resistance may inform new therapeutic strategies that concurrently address multiple cell axes and/or suppressive mechanisms, and clinicians may consequently be able to prioritize targeted therapy combinations for individual patients to improve overall efficacy and outcomes.
Collapse
Affiliation(s)
- Kristin G Anderson
- Department of Microbiology, Immunology and Cancer Biology, Obstetrics and Gynecology, Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | - David A Braun
- Center of Molecular and Cellular Oncology, Yale University Yale Cancer Center, New Haven, Connecticut, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York, USA
| | - Sarah B Gitto
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer L Guerriero
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Brendan Horton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Bridget P Keenan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, USA
| | - Teresa S Kim
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Abigail Overacre-Delgoffe
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Marco Ruella
- Department of Medicine, Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Todd A Triplett
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas, USA
| | - Omkara Veeranki
- Medical Affairs and Clinical Development, Caris Life Sciences Inc, Irving, Texas, USA
| | - Vivek Verma
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Fan Zhang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
29
|
Crosby BJ, Newton RU, Galvão DA, Taaffe DR, Lopez P, Meniawy TM, Khattak MA, Lam WS, Gray ES, Singh F. Feasibility of supervised telehealth exercise for patients with advanced melanoma receiving checkpoint inhibitor therapy. Cancer Med 2023. [PMID: 37184115 DOI: 10.1002/cam4.6091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023] Open
Abstract
PURPOSE To determine the feasibility, safety and preliminary efficacy of a telehealth supervised exercise programme in patients with advanced melanoma receiving checkpoint inhibitor therapy. METHODS A 8-week non-randomised feasibility pilot trial utilising a telehealth delivered multimodal exercise programme undertaken thrice weekly with assessments at baseline and post-intervention. The study was considered feasible if there were no severe or life-threatening adverse events as a result of exercise, and three or more of the following criteria were met: the recruitment rate was >50%, completion rate was >80%, median programme attendance was >75%, median exercise compliance >75%, and average tolerance was >70%. Preliminary efficacy was assessed for objective measures of physical function (2-min step test, repeated chair stand test, 30-s push-up test, and a modified static balance test) and quality of life (QoL), fatigue and other patient-reported outcomes were assessed using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30. RESULTS Eleven patients (32-80 years) were included in the study (6 female, 5 male). The recruitment rate was 48%, completion rate 91%, programme attendance 88%, median exercise compliance 82.1% and 84.9% for resistance and aerobic exercise, respectively, and tolerance 88%, with no severe or life-threatening adverse events as a result of exercise. In terms of preliminary efficacy, physical function significantly improved while QoL was maintained following the intervention. CONCLUSION An 8-week telehealth exercise intervention is feasible and safe for patients with advanced melanoma and appears to improve physical function while preserving QoL during checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Brendan J Crosby
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Robert U Newton
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Human Movement and Nutrition Sciences, University of Queensland, Saint Lucia, Queensland, Australia
| | - Daniel A Galvão
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Dennis R Taaffe
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Pedro Lopez
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia
| | - Tarek M Meniawy
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Muhammad A Khattak
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Wei-Sen Lam
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Favil Singh
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
30
|
Rini BI, Brugarolas J, Atkins MB. Navigating and adapting care integrating immunotherapy, antiangiogenic therapy, and combinations in patients with advanced renal cell carcinoma. J Immunother Cancer 2023; 11:e006361. [PMID: 36898737 PMCID: PMC10008164 DOI: 10.1136/jitc-2022-006361] [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: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Advanced renal cell carcinoma is a biologically heterogeneous disease with multiple treatment options that largely involve immunotherapy and/or anti-angiogenic therapies. The choice of initial and subsequent therapy depends on both clinical and biological considerations. Here, we describe the application of recent data to clinical practice.
Collapse
Affiliation(s)
- Brian I Rini
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James Brugarolas
- University of Texas Southwestern Department of Internal Medicine, Dallas, Texas, USA
| | - Michael B Atkins
- Oncology, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
31
|
Rao D, Lacroix R, Rooker A, Gomes T, Stunnenberg JA, Valenti M, Dimitriadis P, Lin CP, de Bruijn B, Krijgsman O, Ligtenberg MA, Peeper DS, Blank CU. MeVa2.1.dOVA and MeVa2.2.dOVA: two novel BRAFV600E-driven mouse melanoma cell lines to study tumor immune resistance. Melanoma Res 2023; 33:12-26. [PMID: 36545919 DOI: 10.1097/cmr.0000000000000863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
While immunotherapy has become standard-of-care for cutaneous melanoma patients, primary and acquired resistance prevent long-term benefits for about half of the late-stage patients. Pre-clinical models are essential to increase our understanding of the resistance mechanisms of melanomas, aiming to improve the efficacy of immunotherapy. Here, we present two novel syngeneic transplantable murine melanoma cell lines derived from the same primary tumor induced on BrafV600E Pten-/- mice: MeVa2.1 and MeVa2.2. Derivatives of these cell lines expressing the foreign antigen ovalbumin (dOVA) showed contrasting immune-mediated tumor control. MeVa2.2.dOVA melanomas were initially controlled in immune-competent hosts until variants grew out that had lost their antigens. By contrast, MeVa2.1.dOVA tumors were not controlled despite presenting the strong OVA antigen, as well as infiltration of tumor-reactive CD8+ T cells. MeVa2.1.dOVA displayed reduced sensitivity to T cell-mediated killing and growth inhibition in vitro by both IFN-γ and TNF-α. MeVa2.1.dOVA tumors were transiently controlled in vivo by either targeted therapy, adoptive T cell transfer, regulatory T cell depletion, or immune checkpoint blockade. MeVa2.1.dOVA could thus become a valuable melanoma model to evaluate novel immunotherapy combinations aiming to overcome immune resistance mechanisms.
Collapse
Affiliation(s)
- Disha Rao
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Ruben Lacroix
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Alex Rooker
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Tainá Gomes
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Johanna A Stunnenberg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Mesele Valenti
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Petros Dimitriadis
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Chun-Pu Lin
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Beaunelle de Bruijn
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Oscar Krijgsman
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Maarten A Ligtenberg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
| | - Daniel S Peeper
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
- Oncode Institute, Utrecht
| | - Christian U Blank
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Tabolacci C, De Vita D, Facchiano A, Bozzuto G, Beninati S, Failla CM, Di Martile M, Lintas C, Mischiati C, Stringaro A, Del Bufalo D, Facchiano F. Phytochemicals as Immunomodulatory Agents in Melanoma. Int J Mol Sci 2023; 24:2657. [PMID: 36768978 PMCID: PMC9916941 DOI: 10.3390/ijms24032657] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Cutaneous melanoma is an immunogenic highly heterogenic tumor characterized by poor outcomes when it is diagnosed late. Therefore, immunotherapy in combination with other anti-proliferative approaches is among the most effective weapons to control its growth and metastatic dissemination. Recently, a large amount of published reports indicate the interest of researchers and clinicians about plant secondary metabolites as potentially useful therapeutic tools due to their lower presence of side effects coupled with their high potency and efficacy. Published evidence was reported in most cases through in vitro studies but also, with a growing body of evidence, through in vivo investigations. Our aim was, therefore, to review the published studies focused on the most interesting phytochemicals whose immunomodulatory activities and/or mechanisms of actions were demonstrated and applied to melanoma models.
Collapse
Affiliation(s)
- Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Daniela De Vita
- Department of Environmental Biology, University of Rome La Sapienza, 00185 Rome, Italy
| | | | - Giuseppina Bozzuto
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico, 00128 Rome, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, School of Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
33
|
Zob DL, Augustin I, Caba L, Panzaru MC, Popa S, Popa AD, Florea L, Gorduza EV. Genomics and Epigenomics in the Molecular Biology of Melanoma-A Prerequisite for Biomarkers Studies. Int J Mol Sci 2022; 24:ijms24010716. [PMID: 36614156 PMCID: PMC9821083 DOI: 10.3390/ijms24010716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Melanoma is a common and aggressive tumor originating from melanocytes. The increasing incidence of cutaneous melanoma in recent last decades highlights the need for predictive biomarkers studies. Melanoma development is a complex process, involving the interplay of genetic, epigenetic, and environmental factors. Genetic aberrations include BRAF, NRAS, NF1, MAP2K1/MAP2K2, KIT, GNAQ, GNA11, CDKN2A, TERT mutations, and translocations of kinases. Epigenetic alterations involve microRNAs, non-coding RNAs, histones modifications, and abnormal DNA methylations. Genetic aberrations and epigenetic marks are important as biomarkers for the diagnosis, prognosis, and prediction of disease recurrence, and for therapeutic targets. This review summarizes our current knowledge of the genomic and epigenetic changes in melanoma and discusses the latest scientific information.
Collapse
Affiliation(s)
- Daniela Luminita Zob
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, AI. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (I.A.); (L.C.)
| | - Lavinia Caba
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- Correspondence: (I.A.); (L.C.)
| | - Monica-Cristina Panzaru
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Setalia Popa
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Alina Delia Popa
- Nursing Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Laura Florea
- Department of Nephrology-Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| |
Collapse
|
34
|
Vathiotis IA, Salichos L, Martinez-Morilla S, Gavrielatou N, Aung TN, Shafi S, Wong PF, Jessel S, Kluger HM, Syrigos KN, Warren S, Gerstein M, Rimm DL. Baseline gene expression profiling determines long-term benefit to programmed cell death protein 1 axis blockade. NPJ Precis Oncol 2022; 6:92. [PMID: 36522538 PMCID: PMC9755314 DOI: 10.1038/s41698-022-00330-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022] Open
Abstract
Treatment with immune checkpoint inhibitors has altered the course of malignant melanoma, with approximately half of the patients with advanced disease surviving for more than 5 years after diagnosis. Currently, there are no biomarker methods for predicting outcome from immunotherapy. Here, we obtained transcriptomic information from a total of 105 baseline tumor samples comprising two cohorts of patients with advanced melanoma treated with programmed cell death protein 1 (PD-1)-based immunotherapies. Gene expression profiles were correlated with progression-free survival (PFS) within consecutive clinical benefit intervals (i.e., 6, 12, 18, and 24 months). Elastic net binomial regression models with cross validation were utilized to compare the predictive value of distinct genes across time. Lasso regression was used to generate a signature predicting long-term benefit (LTB), defined as patients who remain alive and free of disease progression at 24 months post treatment initiation. We show that baseline gene expression profiles were consistently able to predict long-term immunotherapy outcomes with high accuracy. The predictive value of different genes fluctuated across consecutive clinical benefit intervals, with a distinct set of genes defining benefit at 24 months compared to earlier outcomes. A 12-gene signature was able to predict LTB following anti-PD-1 therapy with an area under the curve (AUC) equal to 0.92 and 0.74 in the training and validation set, respectively. Evaluation of LTB, via a unique signature may complement objective response classification and characterize the logistics of sustained antitumor immune responses.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| | - Leonidas Salichos
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Biological and Chemical Sciences, New York Institute of Technology, New York, USA
| | - Sandra Martinez-Morilla
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Niki Gavrielatou
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Thazin Nwe Aung
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Saba Shafi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Pok Fai Wong
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Shlomit Jessel
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Harriet M Kluger
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Konstantinos N Syrigos
- Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | | | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
35
|
Virgilio T, Bordini J, Cascione L, Sartori G, Latino I, Molina Romero D, Leoni C, Akhmedov M, Rinaldi A, Arribas AJ, Morone D, Seyed Jafari SM, Bersudsky M, Ottolenghi A, Kwee I, Chiaravalli AM, Sessa F, Hunger RE, Bruno A, Mortara L, Voronov E, Monticelli S, Apte RN, Bertoni F, Gonzalez SF. Subcapsular Sinus Macrophages Promote Melanoma Metastasis to the Sentinel Lymph Nodes via an IL1α-STAT3 Axis. Cancer Immunol Res 2022; 10:1525-1541. [PMID: 36206577 PMCID: PMC9716256 DOI: 10.1158/2326-6066.cir-22-0225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/18/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
During melanoma metastasis, tumor cells originating in the skin migrate via lymphatic vessels to the sentinel lymph node (sLN). This process facilitates tumor cell spread across the body. Here, we characterized the innate inflammatory response to melanoma in the metastatic microenvironment of the sLN. We found that macrophages located in the subcapsular sinus (SS) produced protumoral IL1α after recognition of tumoral antigens. Moreover, we confirmed that the elimination of LN macrophages or the administration of an IL1α-specific blocking antibody reduced metastatic spread. To understand the mechanism of action of IL1α in the context of the sLN microenvironment, we applied single-cell RNA sequencing to microdissected metastases obtained from animals treated with the IL1α-specific blocking antibody. Among the different pathways affected, we identified STAT3 as one of the main targets of IL1α signaling in metastatic tumor cells. Moreover, we found that the antitumoral effect of the anti-IL1α was not mediated by lymphocytes because Il1r1 knockout mice did not show significant differences in metastasis growth. Finally, we found a synergistic antimetastatic effect of the combination of IL1α blockade and STAT3 inhibition with stattic, highlighting a new immunotherapy approach to preventing melanoma metastasis.
Collapse
Affiliation(s)
- Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Joy Bordini
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,GenomSys SA, Lugano, Switzerland
| | - Luciano Cascione
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Giulio Sartori
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Daniel Molina Romero
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristina Leoni
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Murodzhon Akhmedov
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alberto J. Arribas
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - S. Morteza Seyed Jafari
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aner Ottolenghi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ivo Kwee
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Anna Maria Chiaravalli
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Robert E. Hunger
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy.,Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Silvia Monticelli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Francesco Bertoni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.,Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Santiago F. Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Corresponding Author: Santiago F. Gonzalez, Institute for Research in Biomedicine, via Francesco Chiesa 5. CH-6500 Bellinzona. Switzerland. Phone: +41 58 666 7226; E-mail:
| |
Collapse
|
36
|
Conway JW, Braden J, Wilmott JS, Scolyer RA, Long GV, Pires da Silva I. The effect of organ-specific tumor microenvironments on response patterns to immunotherapy. Front Immunol 2022; 13:1030147. [DOI: 10.3389/fimmu.2022.1030147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitors, have become widely used in various settings across many different cancer types in recent years. Whilst patients are often treated on the basis of the primary cancer type and clinical stage, recent studies have highlighted disparity in response to immune checkpoint inhibitors at different sites of metastasis, and their impact on overall response and survival. Studies exploring the tumor immune microenvironment at different organ sites have provided insights into the immune-related mechanisms behind organ-specific patterns of response to immunotherapy. In this review, we aimed to highlight the key learnings from clinical studies across various cancers including melanoma, lung cancer, renal cell carcinoma, colorectal cancer, breast cancer and others, assessing the association of site of metastasis and response to immune checkpoint inhibitors. We also summarize the key clinical and pre-clinical findings from studies exploring the immune microenvironment of specific sites of metastasis. Ultimately, further characterization of the tumor immune microenvironment at different metastatic sites, and understanding the biological drivers of these differences, may identify organ-specific mechanisms of resistance, which will lead to more personalized treatment approaches for patients with innate or acquired resistance to immunotherapy.
Collapse
|
37
|
Paris A, Tardif N, Baietti FM, Berra C, Leclair HM, Leucci E, Galibert M, Corre S. The AhR-SRC axis as a therapeutic vulnerability in BRAFi-resistant melanoma. EMBO Mol Med 2022; 14:e15677. [PMID: 36305167 PMCID: PMC9728058 DOI: 10.15252/emmm.202215677] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 12/14/2022] Open
Abstract
The nongenetic mechanisms required to control tumor phenotypic plasticity and shape drug-resistance remain unclear. We show here that the Aryl hydrocarbon Receptor (AhR) transcription factor directly regulates the gene expression program associated with the acquisition of resistance to BRAF inhibitor (BRAFi) in melanoma. In addition, we show in melanoma cells that canonical activation of AhR mediates the activation of the SRC pathway and promotes the acquisition of an invasive and aggressive resistant phenotype to front-line BRAFi treatment in melanoma. This nongenetic reprogramming identifies a clinically compatible approach to reverse BRAFi resistance in melanoma. Using a preclinical BRAFi-resistant PDX melanoma model, we demonstrate that SRC inhibition with dasatinib significantly re-sensitizes melanoma cells to BRAFi. Together we identify the AhR/SRC axis as a new therapeutic vulnerability to trigger resistance and warrant the introduction of SRC inhibitors during the course of the treatment in combination with front-line therapeutics to delay BRAFi resistance.
Collapse
Affiliation(s)
- Anaïs Paris
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Nina Tardif
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Francesca M Baietti
- Laboratory for RNA Cancer Biology, Department of OncologyLKI, KU LeuvenLeuvenBelgium,Trace PDX Platform, Department of OncologyLKI, KU LeuvenLeuvenBelgium
| | - Cyrille Berra
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance,Department of Molecular Genetics and GenomicsHospital University of Rennes (CHU Rennes)RennesFrance
| | - Héloïse M Leclair
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of OncologyLKI, KU LeuvenLeuvenBelgium,Trace PDX Platform, Department of OncologyLKI, KU LeuvenLeuvenBelgium
| | - Marie‐Dominique Galibert
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance,Department of Molecular Genetics and GenomicsHospital University of Rennes (CHU Rennes)RennesFrance
| | - Sébastien Corre
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) – UMR6290, ERL U1305RennesFrance
| |
Collapse
|
38
|
Lopes J, Rodrigues CMP, Gaspar MM, Reis CP. Melanoma Management: From Epidemiology to Treatment and Latest Advances. Cancers (Basel) 2022; 14:4652. [PMID: 36230575 PMCID: PMC9562203 DOI: 10.3390/cancers14194652] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is the deadliest skin cancer, whose morbidity and mortality indicators show an increasing trend worldwide. In addition to its great heterogeneity, melanoma has a high metastatic potential, resulting in very limited response to therapies currently available, which were restricted to surgery, radiotherapy and chemotherapy for many years. Advances in knowledge about the pathophysiological mechanisms of the disease have allowed the development of new therapeutic classes, such as immune checkpoint and small molecule kinase inhibitors. However, despite the incontestable progress in the quality of life and survival rates of the patients, effectiveness is still far from desired. Some adverse side effects and resistance mechanisms are the main barriers. Thus, the search for better options has resulted in many clinical trials that are now investigating new drugs and/or combinations. The low water solubility of drugs, low stability and rapid metabolism limit the clinical potential and therapeutic use of some compounds. Thus, the research of nanotechnology-based strategies is being explored as the basis for the broad application of different types of nanosystems in the treatment of melanoma. Future development focus on challenges understanding the mechanisms that make these nanosystems more effective.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Pinto Reis
- Research Institute for Medicines, iMed.ULisboa—Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
- Instituto de Biofísica e Engenharia Biomédica, IBEB, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
39
|
Vos WG, Lutgens E, Seijkens TTP. Statins and immune checkpoint inhibitors: a strategy to improve the efficacy of immunotherapy for cancer? J Immunother Cancer 2022. [PMCID: PMC9442479 DOI: 10.1136/jitc-2022-005611] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the past decade, immune checkpoint inhibitor (ICI) therapy significantly improved the prognosis of patients with cancer. Despite impressive and often unprecedented response rates, a significant portion of the patients fails to benefit from this treatment. Additional strategies to improve ICI efficacy are therefore needed. The widespread clinical use of ICIs has increased our knowledge on the effects of the concomitant use of commonly prescribed drugs on the outcome of ICI treatment. A particular interesting class of drugs in this context are statins. These HMG-CoA reductase inhibitors, which are used to treat hypercholesterolemia and reduce the risk for atherosclerotic cardiovascular disease, are frequently used by patients with (advanced) cancer. This paper addresses the hypothesis that statins improve the efficacy of ICI therapy.
Collapse
Affiliation(s)
- Winnie G Vos
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Lutgens
- Cardiovascular Medicine, Mayo Clinic, Rochester, New York, USA
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universitat Munchen, Munchen, Germany
| | - Tom T P Seijkens
- Medical Biochemistry, Amsterdam University Medical Centres, Duivendrecht, The Netherlands
- Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Epshtein Y, Blau R, Pisarevsky E, Koshrovski-Michael S, Ben-Shushan D, Pozzi S, Shenbach-Koltin G, Fridrich L, Buzhor M, Krivitsky A, Dey P, Satchi-Fainaro R. Polyglutamate-based nanoconjugates for image-guided surgery and post-operative melanoma metastases prevention. Theranostics 2022; 12:6339-6362. [PMID: 36168618 PMCID: PMC9475454 DOI: 10.7150/thno.72941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/21/2022] [Indexed: 01/01/2023] Open
Abstract
Rationale: Cutaneous melanoma is the most aggressive and deadliest of all skin malignancies. Complete primary tumor removal augmented by advanced imaging tools and effective post-operative treatment is critical in the prevention of tumor recurrence and future metastases formation. Methods: To meet this challenge, we designed novel polymeric imaging and therapeutic systems, implemented in a two-step theranostic approach. Both are composed of the biocompatible and biodegradable poly(α,L-glutamic acid) (PGA) nanocarrier that facilitates extravasation-dependent tumor targeting delivery. The first system is a novel, fluorescent, Turn-ON diagnostic probe evaluated for the precise excision of the primary tumor during image-guided surgery (IGS). The fluorescence activation of the probe occurs via PGA degradation by tumor-overexpressed cathepsins that leads to the separation of closely-packed, quenched FRET pair. This results in the emission of a strong fluorescence signal enabling the delineation of the tumor boundaries. Second, therapeutic step is aimed to prevent metastases formation with minimal side effects and maximal efficacy. To that end, a targeted treatment containing a BRAF (Dabrafenib - mDBF)/MEK (Selumetinib - SLM) inhibitors combined on one polymeric platform (PGA-SLM-mDBF) was evaluated for its anti-metastatic, preventive activity in combination with immune checkpoint inhibitors (ICPi) αPD1 and αCTLA4. Results: IGS in melanoma-bearing mice led to a high tumor-to-background ratio and reduced tumor recurrence in comparison with mice that underwent surgery under white light (23% versus 33%, respectively). Adjuvant therapy with PGA-SLM-mDBF combined with ICPi, was well-tolerated and resulted in prolonged survival and prevention of peritoneal and brain metastases formation in BRAF-mutated melanoma-bearing mice. Conclusions: The results reveal the great clinical potential of our PGA-based nanosystems as a tool for holistic melanoma treatment management.
Collapse
Affiliation(s)
- Yana Epshtein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rachel Blau
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, Mail Code 0448, La Jolla, CA 92093-0448
| | - Evgeni Pisarevsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lidar Fridrich
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Buzhor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
41
|
Lin EPY, Hsu CY, Berry L, Bunn P, Shyr Y. Analysis of Cancer Survival Associated With Immune Checkpoint Inhibitors After Statistical Adjustment: A Systematic Review and Meta-analyses. JAMA Netw Open 2022; 5:e2227211. [PMID: 35976648 PMCID: PMC9386543 DOI: 10.1001/jamanetworkopen.2022.27211] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
IMPORTANCE Appropriate clinical decision-making relies on accurate data interpretation, which in turn relies on the use of suitable statistical models. Long tails and early crossover-2 features commonly observed in immune checkpoint inhibitor (ICI) survival curves-raise questions as to the suitability of Cox proportional hazards regression for ICI survival analysis. Cox proportional hazards-Taylor expansion adjustment for long-term survival data (Cox-TEL) adjustment may provide possible solutions in this setting. OBJECTIVE To estimate overall survival and progression-free survival benefits of ICI therapy vs chemotherapy using Cox-TEL adjustment. DATA SOURCES A PubMed search was performed for all cataloged publications through May 22, 2022. STUDY SELECTION The search was restricted to randomized clinical trials with search terms for ICIs and lung cancer, melanoma, or urothelial carcinoma. The publications identified were further reviewed for inclusion. DATA EXTRACTION AND SYNTHESIS Cox proportional hazards ratios (HRs) were transformed to Cox-TEL HRs for patients with short-term treatment response (ie, short-term survivor) (ST-HR) and difference in proportions for patients with long-term survival (LT-DP) by Cox-TEL. Meta-analyses were performed using a frequentist random-effects model. MAIN OUTCOMES AND MEASURES Outcomes of interest were pooled overall survival (primary outcome) and progression-free survival (secondary outcome) HRs, ST-HRs, and LT-DPs. Subgroup analyses stratified by cancer type also were performed. RESULTS A total of 1036 publications was identified. After 3 levels of review against inclusion criteria, 13 clinical trials (7 in non-small cell lung cancer, 3 in melanoma, and 3 in urothelial carcinoma) were selected for the meta-analysis. In the primary analysis, pooled findings were 0.75 (95% CI, 0.70-0.81) for HR, 0.86 (95% CI, 0.81-0.92) for ST-HR, and 0.08 (95% CI, 0.06-0.10) for LT-DP. In the secondary analysis, the pooled values for progression-free survival were 0.77 (95% CI, 0.64-0.91) for HR, 1.02 (95% CI, 0.84-1.24) for ST-HR, and 0.10 (95% CI, 0.06-0.14) for LT-DP. CONCLUSIONS AND RELEVANCE This systematic review and meta-analysis of ICI clinical trial results noted consistently larger ST-HRs vs Cox HRs for ICI therapy, with an LT-DP of approximately 10%. These results suggest that Cox HRs may not provide a full picture of survival outcomes when the risk reduction from treatment is not constant, which may aid in the decision-making process of oncologists and patients.
Collapse
Affiliation(s)
- Emily Pei-Ying Lin
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Yuan Hsu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne Berry
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul Bunn
- Department of Medicine, University of Colorado School of Medicine, Aurora
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
42
|
Pala L, Sala I, Oriecuia C, De Pas T, Queirolo P, Specchia C, Cocorocchio E, Ferrucci P, Patanè D, Saponara M, Pennacchioli E, Coppola S, Viale G, Giaccone G, Gelber RD, Bagnardi V, Conforti F. Association of Anticancer Immune Checkpoint Inhibitors With Patient-Reported Outcomes Assessed in Randomized Clinical Trials: A Systematic Review and Meta-analysis. JAMA Netw Open 2022; 5:e2226252. [PMID: 35972744 PMCID: PMC9382448 DOI: 10.1001/jamanetworkopen.2022.26252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Importance The association of immune checkpoint inhibitors (ICIs) with patient quality of life has been poorly explored. Objective To evaluate patient-reported outcomes (PROs) assessed in randomized clinical trials (RCTs) of immunotherapy-based treatments. Data Sources This systematic review and random-effects meta-analysis used RCTs identified in PubMed, MEDLINE, Embase, and Scopus from database inception to June 1, 2021. Study Selection A total of 2259 RCTs were identified that assessed ICIs as monotherapy or in combination with chemotherapy or combined with another ICI and/or targeted therapy vs control groups not containing immunotherapy in patients with advanced solid tumors. Studies were reviewed independently by 2 authors. Data Extraction and Synthesis This meta-analysis followed the PRISMA guidelines and recommendations of the Setting International Standards in Analyzing Patient-Reported Outcomes and Quality of Life Endpoints Data Consortium. Main Outcomes and Measures The coprimary aims of the meta-analysis were (1) pooled differences between treatment groups in the mean change of PRO score from baseline to 12 and 24 weeks of follow-up and (2) pooled differences between treatment groups in the time to deterioration of PRO score. For each end point, RCTs have been analyzed according to the type of treatment administered in the experimental group: ICIs given as monotherapy, ICIs combined with chemotherapy, or ICIs in association with another ICI and/or with targeted therapies. Results Of the 2259 identified RCTs, 34 (18 709 patients) met the selection criteria and were analyzed. In the group of 19 RCTs testing ICIs as monotherapy, the pooled between-groups difference of mean change from baseline to 12 weeks of follow-up was 4.6 (95% CI, 2.8-6.4), and the mean change from baseline to 24 weeks of follow-up was 6.1 (95% CI, 4.2-8.1), significantly favoring ICIs. The pooled difference was 1.4 (95% CI, -0.4 to 3.2) at week 12 and 2.5 (95% CI, -0.8 to 5.9) at week 24 in the group of 8 RCTs testing ICIs combined with chemotherapy and 2.1 (95% CI, -0.8 to 5.0) at week 12 and 2.1 (95% CI, -0.4 to 4.5) at week 24 in the group of 8 RCTs testing other ICI-containing combinations. The time to deterioration was significantly longer in the immunotherapy-containing groups compared with control groups in all 3 groups of RCTs evaluated (hazard ratios of 0.80 [95% CI, 0.70-0.91] for ICIs as monotherapy, 0.89 [95% CI, 0.78-1.00] for ICIs plus chemotherapy, and 0.78 [95% CI, 0.63-0.96] for other ICI-containing combinations). Conclusions and Relevance Immune checkpoint inhibitors as monotherapy appear to have a favorable association with patient-reported quality of life and can be combined with other classes of anticancer drugs without worsening this quality of life.
Collapse
Affiliation(s)
- Laura Pala
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
- Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Isabella Sala
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Chiara Oriecuia
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Tommaso De Pas
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
- Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Paola Queirolo
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emilia Cocorocchio
- Division of Medical Oncology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Pierfrancesco Ferrucci
- Department of Experimental Oncology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Damiano Patanè
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
| | - Maristella Saponara
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
| | - Elisabetta Pennacchioli
- Melanoma, Sarcoma, and Rare Tumors Surgery Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Sara Coppola
- Melanoma, Sarcoma, and Rare Tumors Surgery Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | | | - Richard D. Gelber
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Frontier Science & Technology Research Foundation, Boston, Massachusetts
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Fabio Conforti
- Division of Melanoma, Sarcomas, and Rare Tumors, European Institute of Oncology, Milan, Italy
- Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| |
Collapse
|
43
|
Papak I, Chruściel E, Dziubek K, Kurkowiak M, Urban-Wójciuk Z, Marjański T, Rzyman W, Marek-Trzonkowska N. What Inhibits Natural Killers’ Performance in Tumour. Int J Mol Sci 2022; 23:ijms23137030. [PMID: 35806034 PMCID: PMC9266640 DOI: 10.3390/ijms23137030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 12/21/2022] Open
Abstract
Natural killer cells are innate lymphocytes with the ability to lyse tumour cells depending on the balance of their activating and inhibiting receptors. Growing numbers of clinical trials show promising results of NK cell-based immunotherapies. Unlike T cells, NK cells can lyse tumour cells independent of antigen presentation, based simply on their activation and inhibition receptors. Various strategies to improve NK cell-based therapies are being developed, all with one goal: to shift the balance to activation. In this review, we discuss the current understanding of ways NK cells can lyse tumour cells and all the inhibitory signals stopping their cytotoxic potential.
Collapse
Affiliation(s)
- Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Elżbieta Chruściel
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Zuzanna Urban-Wójciuk
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Tomasz Marjański
- Department of Thoracic Surgery, Medical University of Gdansk, 80-210 Gdansk, Poland; (T.M.); (W.R.)
| | - Witold Rzyman
- Department of Thoracic Surgery, Medical University of Gdansk, 80-210 Gdansk, Poland; (T.M.); (W.R.)
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence:
| |
Collapse
|
44
|
Austin E, Huang A, Wang JY, Cohen M, Heilman E, Maverakis E, Michl J, Jagdeo J. Red Light Phototherapy Using Light-Emitting Diodes Inhibits Melanoma Proliferation and Alters Tumor Microenvironments. Front Oncol 2022; 12:928484. [PMID: 35847848 PMCID: PMC9278815 DOI: 10.3389/fonc.2022.928484] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 01/26/2023] Open
Abstract
Background Total annual cancer rates have decreased due to improved treatment and prevention. However, the incidence of melanoma is rising, and not all patients respond to immune and targeted approaches. Therefore, we sought to determine the efficacy of red light (RL) phototherapy in preclinical models of melanoma. Methods Melanoma cells (A375, B16F10, MNT-1) were irradiated with RL. Melanoma proliferation, apoptosis, oxidative stress, and p53 phosphorylation were measured in vitro. In C57BL/6 mice, phototherapy safety, B16F10 tumor growth, and immunocyte infiltration were assessed following RL. Results In vitro, 640 J/cm2 RL decreased cellular proliferation without increasing apoptosis, while 1280 J/cm2 increased apoptosis. RL increased intracellular reactive oxygen species generation and p53 phosphorylation. In animal models, 2560 J/cm2 RL significantly prevented melanoma growth and increased the expression of CD103+ dendritic cells. 1280 and 1920 J/cm2 RL decreased tumor volume, but not significantly. RL did not cause skin inflammation or erythema in normal skin. Conclusion RL represents a potentially safe and effective melanoma therapeutic. RL prevented tumor growth and increased the expression of immune markers, such as CD103, that are associated with favorable melanoma outcomes. Further research is needed to determine the optimal clinical treatment regimen for melanoma using RL.
Collapse
Affiliation(s)
- Evan Austin
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States,Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States
| | - Alisen Huang
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Jennifer Y. Wang
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Marc Cohen
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Edward Heilman
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States
| | - Emanual Maverakis
- Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States
| | - Josef Michl
- Department of Pathology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Jared Jagdeo
- Department of Dermatology, State University of New York (SUNY) Downstate Medical Center, Brooklyn, NY, United States,Department of Dermatology, University of California (UC) Davis Medical Center, Sacramento, CA, United States,*Correspondence: Jared Jagdeo,
| |
Collapse
|
45
|
Navani V, Graves MC, Mandaliya H, Hong M, van der Westhuizen A, Martin J, Bowden NA. Melanoma: An immunotherapy journey from bench to bedside. Cancer Treat Res 2022; 183:49-89. [PMID: 35551656 DOI: 10.1007/978-3-030-96376-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Melanoma gave science a window into the role immune evasion plays in the development of malignancy. The entire spectrum of immune focused anti-cancer therapies has been subjected to clinical trials in this disease, with limited success until the immune checkpoint blockade era. That revolution launched first in melanoma, heralded a landscape change throughout cancer that continues to reverberate today.
Collapse
Affiliation(s)
| | - Moira C Graves
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia
| | - Hiren Mandaliya
- Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Martin Hong
- Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Andre van der Westhuizen
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia.,Calvary Mater Hospital Newcastle, Edith St, Waratah, NSW, 2298, Australia
| | - Jennifer Martin
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia.,John Hunter Hospital, Newcastle, NSW, Australia
| | - Nikola A Bowden
- Centre for Drug Repurposing and Medicines Research, University of Newcastle and Hunter Medical Research Institute, University Dr, Callaghan, NSW, 2308, Australia
| |
Collapse
|
46
|
Proton Pump Inhibitor Use and Efficacy of Nivolumab and Ipilimumab in Advanced Melanoma. Cancers (Basel) 2022; 14:cancers14092300. [PMID: 35565428 PMCID: PMC9103038 DOI: 10.3390/cancers14092300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Immune checkpoint inhibitors have been shown to improve survival in patients with advanced melanoma; however, a proportion of patients do not experience durable clinical benefit with these agents. Findings from a previous study suggested that the use of proton pump inhibitors while receiving immune checkpoint inhibitors may lead to worse clinical outcomes. To validate those results, we performed this retrospective analysis using data from three clinical trials involving patients with advanced melanoma treated with immune checkpoint inhibitors. We found that there is not enough evidence to conclude that proton pump inhibitors influence the efficacy of immune checkpoint inhibitors. Prospective studies are needed to conclusively determine if the use of proton pump inhibitors has any meaningful impact on the efficacy of immune checkpoint inhibitors in patients with advanced melanoma. Abstract The impact of proton pump inhibitors (PPIs) on clinical outcomes with first-line immune checkpoint inhibitors (ICIs) in patients with metastatic melanoma was previously analyzed in the phase II study, CheckMate 069. This retrospective analysis utilized data from three phase II/III studies of first-line ICI therapy in untreated advanced melanoma: CheckMate 066, 067, and 069. All randomized patients with PPI use ≤ 30 days before initiating study treatment were included in the PPI-use subgroup. Possible associations between baseline PPI use and efficacy were evaluated within each treatment arm of each study using multivariable modeling. Approximately 20% of 1505 randomized patients across the studies reported baseline PPI use. The median follow-up was 52.6–58.5 months. Objective response rate (ORR), progression-free survival (PFS), and overall survival analyses provided insufficient evidence of a meaningful association between PPI use and efficacy outcomes with nivolumab-plus-ipilimumab, nivolumab, or ipilimumab therapy. In five of the six ICI treatment arms, 95% confidence intervals for odds ratios or hazard ratios traversed 1. Significant associations were observed in the CheckMate 069 combination arm between PPI use and poorer ORR and PFS. This multivariable analysis found insufficient evidence to support meaningful associations between PPI use and ICI efficacy in patients with advanced melanoma.
Collapse
|
47
|
Atkins MB, Julian C, Secrest MH, Lee J, Abajo-Guijarro AM, McKenna E. Real-world treatment patterns and overall survival in BRAF-mutant melanoma patients treated with immunotherapy or targeted therapy. Future Oncol 2022; 18:2233-2245. [PMID: 35441522 DOI: 10.2217/fon-2021-1536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To assess overall survival (OS) in patients with advanced BRAF-mutant melanoma by first-line (1L) targeted therapy (TT) or checkpoint inhibitor (CPI) use, second-line (2L) TT or CPI use, and treatment sequence. Patients & methods: Advanced BRAF-mutant melanoma patients treated with 1L CPI or TT were selected from a real-world, electronic health record-derived database. Results: CPI was associated with improved survival after adjustment for potential confounders (hazard ratio, 0.75 [95% CI, 0.66-0.87]). Median OS was similar between 2L therapies and among likely treatment sequences. Conclusion: This real-world study demonstrated a survival benefit with 1L CPI versus TT. Analyses of 2L and treatment sequences were unable to detect or rule out clinically relevant differences in OS.
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | | | | | - Janet Lee
- Genentech, Inc, South San Francisco, CA 94080, USA
| | | | | |
Collapse
|
48
|
Knochelmann HM, Ware MB, Rali A, Linderman S, Shantha JG, Lawson DH, Yushak M, Swerlick R, Paulos CM, Yeh S, Kudchadkar R. Case Report: Delayed Onset Multi-Organ Toxicities in a Melanoma Patient Achieving Complete Response to BRAF/MEK Inhibition. Front Oncol 2022; 12:836845. [PMID: 35433480 PMCID: PMC9008700 DOI: 10.3389/fonc.2022.836845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 01/11/2023] Open
Abstract
Autoimmune toxicities, while common following treatment with cancer immunotherapies, are not well-characterized in patients treated with BRAF/MEK inhibitors. Emerging data suggest that autoimmune effects may be linked with superior responses to both treatment modalities; however, there is little evidence describing mechanisms of immune-related toxicity for patients on BRAF/MEK inhibitors. Here we describe the experience of a 59-year-old HLA-A2, A29, B27-positive male with recurrent/metastatic melanoma. After progression on checkpoint inhibitor therapy, he was treated with dabrafenib/trametinib followed by encorafenib/binimetinib, which were well-tolerated and resulted in a complete response. Eighteen months into BRAF/MEK inhibitor therapy, and three months after initially finding a complete response, he developed a series of sudden-onset, severe toxicities: namely, bilateral panuveitis, cytopenias, joint pain, skin rash, hypercalcemia, and interstitial nephritis, which led to BRAF/MEKi cessation. Immunological analyses revealed induction of a peripheral type-17 cytokine signature characterized by high IL-23, IL-6, IL-10, IL-17A/F, IL-1β, and IL-21 among other cytokines in plasma corresponding with the height of symptoms. These findings highlight a novel instance of delayed autoimmune-like reaction to BRAF/MEK inhibition and identify a possible role for Th/Tc17 activation in their pathogenesis thus warranting future clinical and immunological characterization.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery: Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Michael Brandon Ware
- Department of Surgery: Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Aditya Rali
- Emory Eye Center, Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, United States
| | - Susanne Linderman
- Emory Eye Center, Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, United States.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Jessica G Shantha
- Emory Eye Center, Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, United States
| | - David H Lawson
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Melinda Yushak
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Robert Swerlick
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, United States
| | - Chrystal M Paulos
- Department of Surgery: Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Steven Yeh
- Emory Eye Center, Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, United States.,Truhlsen Eye Institute, Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Ragini Kudchadkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| |
Collapse
|
49
|
Real-World Therapy with Pembrolizumab: Outcomes and Surrogate Endpoints for Predicting Survival in Advanced Melanoma Patients in Germany. Cancers (Basel) 2022; 14:cancers14071804. [PMID: 35406577 PMCID: PMC8997941 DOI: 10.3390/cancers14071804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Knowledge on the real-world outcomes of patients with advanced melanoma and the value of different endpoints for evaluating survival benefits is limited. We investigated the outcomes and different surrogate endpoints for overall survival (OS) in 664 pembrolizumab-treated patients with advanced melanoma in Germany. Our findings support the effectiveness of pembrolizumab in real-world clinical practice. The real-world time to next treatment was most strongly correlated with OS, suggesting it as a valuable surrogate endpoint to assess treatment effectiveness. Real-world studies assessing time to next treatment could support clinical and payer decision making. Abstract Knowledge on the real-world characteristics and outcomes of pembrolizumab-treated advanced melanoma patients in Germany and on the value of different real-world endpoints as surrogates for overall survival (OS) is limited. A sample of 664 pembrolizumab-treated patients with advanced melanoma from the German registry ADOReg was used. We examined OS, real-world progression-free survival (rwPFS), real-world time to next treatment (rwTtNT), and real-world time on treatment (rwToT). Spearman’s rank and iterative multiple imputation (IMI)-based correlation coefficients were computed between the OS and the rwPFS, rwTtNT, and rwToT and reported for the first line of therapy and the overall sample. The median OS was 30.5 (95%CI 25.0–35.4) months, the rwPFS was 3.9 months (95%CI 3.5–4.9), the rwTtNT was 10.7 months (95%CI 9.0–12.9), and the rwToT was 6.2 months (95%CI 5.1–6.8). The rwTtNT showed the highest correlation with the OS based on the IMI (rIMI = 0.83), Spearman rank correlations (rs = 0.74), followed by the rwToT (rIMI = 0.74 and rs = 0.65) and rwPFS (rIMI = 0.69 and rs = 0.56). The estimates for the outcomes and correlations were similar for the overall sample and those in first-line therapy. The median OS was higher compared to recent real-world studies, supporting the effectiveness of pembrolizumab in regular clinical practice. The rwTtNT may be a valuable OS surrogate, considering the highest correlation was observed with the OS among the investigated real-world endpoints.
Collapse
|
50
|
Arteaga Ceballos DP, Saeed-Kamil Z, King I, Stockley T, Liu D, Muniz TP, Saibil SD, Hogg D, Spreafico A, Butler MO. Turnaround Times in Melanoma BRAF Testing and the Impact on the Initiation of Systemic Therapy at a Single Tertiary Care Cancer Center. JCO Oncol Pract 2022; 18:e642-e647. [PMID: 35363503 DOI: 10.1200/op.21.00810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The identification of BRAF mutations in melanoma enables targeted therapy and improves patient outcomes. Barriers to BRAF molecular testing affect the quality of care and therapeutic options. METHODS This retrospective study mapped BRAF testing timelines in adult patients with melanoma at the Princess Margaret Cancer Centre to identify obstacles to timely BRAF reporting and its impact on the initiation of therapy. RESULTS Sixty-six cases were included. The median time between BRAF request and result was 12 days (95% CI, 8 to 15) when the BRAF test was ordered by pathology, compared with 20 days (95% CI, 16 to 23) if the test was requested by another specialist (P < .001). When the BRAF test and biopsy were performed within the same institution, the BRAF median turnaround time (TAT) was 13 days (95% CI, 6 to 19) compared with 19 days (95% CI, 16 to 21) if the sample was transferred from another institution (P = .02). Forty-seven patients received systemic therapy, and 20 had metastatic disease. In the metastatic subgroup, if the BRAF result was available at the first medical oncology visit, the initiation of treatment was 20 days (95% CI, 9.6 to 30.3), but was delayed to 31 days (95% CI, 10.8 to 51.1) if the BRAF result was not available (P = .03). CONCLUSION This study showed variations in BRAF test results in TAT. One factor affecting this timeline is the transfer time, which can be streamlined by pathology reflex testing. Delays in TAT affect the timing and type of therapeutic intervention, especially in patients with stage IV disease.
Collapse
Affiliation(s)
- Diana Paola Arteaga Ceballos
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada
| | - Zaid Saeed-Kamil
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ian King
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Tracy Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Diane Liu
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Thiago P Muniz
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada
| | - Samuel D Saibil
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada.,Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - David Hogg
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada
| | - Anna Spreafico
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada.,Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Marcus O Butler
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Canada.,Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|