401
|
Low SSP, El-Shakankery K, Brown E, Christie A, McCormack S, Stares M. Case report: A rare case of immunotherapy induced isolated left CN VI palsy in a patient with unresectable melanoma. Front Oncol 2024; 14:1330271. [PMID: 38410107 PMCID: PMC10896602 DOI: 10.3389/fonc.2024.1330271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction Immune checkpoint inhibitors are the mainstay of treatment in patients with unresectable or metastatic melanoma. Combination immunotherapy with ipilimumab and nivolumab has shown to improve survival outcomes as compared to single agent immunotherapy in these patients. Neurological immune-related adverse effects (irAEs) are uncommon and cranial nerve palsies are seen even more infrequently. Case presentation A 66-year-old woman with a background of metastatic, unresectable melanoma with supraclavicular and axillary lymph nodal involvement presented with a headache, photophobia and diplopia 3 weeks after her first cycle of ipilimumab and nivolumab. She was subsequently diagnosed with a left-sided cranial nerve VI palsy and treated with high dose oral steroids and steroid eye drops, with complete resolution of symptoms. She also experienced Grade 3 dermatitis requiring topical steroids, Grade 2 hypothyroidism and vitiligo. She continues to have an excellent clinical and radiological response, despite further immunotherapy being suspended. Conclusion This is the first reported UK case of immunotherapy-induced isolated cranial nerve VI palsy. Multiple irAEs are more common with combination immunotherapy and its occurrence is associated with more favourable outcomes in melanoma. Immunotherapy continues to revolutionise oncological care, but clinicians must be cognizant of unpredictable irAEs, which may require prompt assessment and intervention.
Collapse
Affiliation(s)
- Samantha Su Ping Low
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | - Karim El-Shakankery
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | - Ewan Brown
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | - Alan Christie
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | | | - Mark Stares
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
402
|
Hsieh MY, Hsu SK, Liu TY, Wu CY, Chiu CC. Melanoma biology and treatment: a review of novel regulated cell death-based approaches. Cancer Cell Int 2024; 24:63. [PMID: 38336727 PMCID: PMC10858604 DOI: 10.1186/s12935-024-03220-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
The incidence of melanoma, the most lethal form of skin cancer, has increased due to ultraviolet exposure. The treatment of advanced melanoma, particularly metastatic cases, remains challenging with poor outcomes. Targeted therapies involving BRAF/MEK inhibitors and immunotherapy based on anti-PD1/anti-CTLA4 antibodies have achieved long-term survival rates of approximately 50% for patients with advanced melanoma. However, therapy resistance and inadequate treatment response continue to hinder further breakthroughs in treatments that increase survival rates. This review provides an introduction to the molecular-level pathogenesis of melanoma and offers an overview of current treatment options and their limitations. Cells can die by either accidental or regulated cell death (RCD). RCD is an orderly cell death controlled by a variety of macromolecules to maintain the stability of the internal environment. Since the uncontrolled proliferation of tumor cells requires evasion of RCD programs, inducing the RCD of melanoma cells may be a treatment strategy. This review summarizes studies on various types of nonapoptotic RCDs, such as autophagy-dependent cell death, necroptosis, ferroptosis, pyroptosis, and the recently discovered cuproptosis, in the context of melanoma. The relationships between these RCDs and melanoma are examined, and the interplay between these RCDs and immunotherapy or targeted therapy in patients with melanoma is discussed. Given the findings demonstrating melanoma cell death in response to different stimuli associated with these RCDs, the induction of RCD shows promise as an integral component of treatment strategies for melanoma.
Collapse
Affiliation(s)
- Ming-Yun Hsieh
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Tzu-Yu Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
403
|
Attias M, Piccirillo CA. The impact of Foxp3 + regulatory T-cells on CD8 + T-cell dysfunction in tumour microenvironments and responses to immune checkpoint inhibitors. Br J Pharmacol 2024. [PMID: 38325330 DOI: 10.1111/bph.16313] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/23/2023] [Accepted: 01/01/2024] [Indexed: 02/09/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been a breakthrough in cancer therapy, inducing durable remissions in responding patients. However, they are associated with variable outcomes, spanning from disease hyperprogression to complete responses with the onset of immune-related adverse events. The consequences of checkpoint inhibition on Foxp3+ regulatory T (Treg ) cells remain unclear but could provide key insights into these variable outcomes. In this review, we first cover the mechanisms that underlie the development of hot and cold tumour microenvironments, which determine the efficacy of immunotherapy. We then outline how differences in tumour-intrinsic immunogenicity, T-cell trafficking, local metabolic environments and inhibitory checkpoint signalling differentially impair CD8+ T-cell function in tumour microenvironments, all the while promoting Treg -cell suppressive activity. Finally, we focus on the mechanisms that enable the induction of polyfunctional CD8+ T-cells upon checkpoint blockade and discuss the role of ICI-induced Treg -cell reactivation in acquired resistance to treatment.
Collapse
Affiliation(s)
- Mikhaël Attias
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
| |
Collapse
|
404
|
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
|
405
|
Kochanek C, Gilde C, Zimmer L, Ugurel S, Meier F, Utikal J, Pföhler C, Herbst R, Haferkamp S, Welzel J, Dücker P, Leiter U, Weichenthal M, von Wasielewski I, Angela Y, Gutzmer R. Effects of an immunosuppressive therapy on the efficacy of immune checkpoint inhibition in metastatic melanoma - An analysis of the prospective skin cancer registry ADOREG. Eur J Cancer 2024; 198:113508. [PMID: 38183763 DOI: 10.1016/j.ejca.2023.113508] [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: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND The impact of immunosuppressive therapy (IST) on immune-checkpoint inhibition (ICI) is unclear. METHODS Patients with unresectable advanced melanoma (MM) treated with ICI in the years 2011-2020 were identified from the prospective multicenter German skin cancer registry ADOREG. Patients with IST within 60 days before, or within 30 days after start of ICI were compared to patients without IST. End points were disease control rate (DCR), overall survival (OS) and progression-free survival (PFS) determined by Kaplan-Meier method. Prognostic factors were evaluated in a Cox regression model. RESULTS Of 814 patients treated with ICI, 73 (9%) received concomitant IST, mainly steroids. Patients with brain metastases (BM) received IST more frequently (n = 34/130 patients; 26%), than patients without BM (39/684 patients; 6%). In patients without BM, IST initiated before, but not IST initiated after start of ICI was significantly associated with worse PFS (univariate hazard ratio (HR) 2.59, 95% confidence interval (95%-CI) 1.07-6.28, p = 0.035; multivariate HR 3.48, 95%-CI 1.26-9.6, p = 0.016). There was no association between IST and OS or DCR. In patients with BM, IST initiated before, but not after start of ICI was significantly associated with worse OS (univariate HR 2.06, 95%-CI 1.07-3.95, p = 0.031; multivariate HR 5.91, 95%-CI 1.74-20.14, p = 0.004). There was no association between IST and PFS or DCR. CONCLUSION Patients receiving IST 60 days before start of ICI showed a tendency to an impaired therapy outcome. IST initiated within 30 days after start of ICI, mainly due to early side effects, did not affect the efficacy of ICI therapy.
Collapse
Affiliation(s)
- Corinna Kochanek
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany.
| | - Catharina Gilde
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany
| | - Lisa Zimmer
- Department of Dermatology, Venerology and Allergology, University Hospital Essen & Westdeutsches Tumorzentrum, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venerology and Allergology, University Hospital Essen & Westdeutsches Tumorzentrum, Essen, Germany; German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, Dresden, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venerology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector CancerInstitute at the University Medical Center Mannheim, Mannheim, Germany
| | - Claudia Pföhler
- Department of Dermatology, Venerology and Allergology, Skin Cancer Centre, Saarland University Medical School, Homburg, Saar, Germany
| | - Rudolf Herbst
- Department of Dermatology and Allergology, Skin Cancer Centre, Helios Clinic Erfurt, Erfurt, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, Skin Cancer Centre, University Hospital Regensburg, Regensburg, Germany
| | - Julia Welzel
- Department of Dermatology and Allergology, Skin Cancer Centre, Augsburg University Hospital, Augsburg, Germany
| | - Pia Dücker
- Department of Dermatology, Skin Cancer Centre, Hospital Dortmund, Dortmund, Germany
| | - Ulrike Leiter
- Centre of Dermatooncology, Department of Dermatology, Venerology and Allergology, University of Tuebingen, Germany
| | - Michael Weichenthal
- Department of Dermatology, Venerology and Allergology, Skin Cancer Centre, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Imke von Wasielewski
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany
| | - Yenny Angela
- Department of Dermatology, Johannes Wesling Medical Center Minden, Ruhr University Bochum Campus Minden, Minden, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center Minden, Ruhr University Bochum Campus Minden, Minden, Germany
| |
Collapse
|
406
|
Di Giacomo AM, Lahn M, Eggermont AM, Fox B, Ibrahim R, Sharma P, Allison JP, Maio M. The future of targeting cytotoxic T-lymphocyte-associated protein-4: Is there a role? Eur J Cancer 2024; 198:113501. [PMID: 38169219 DOI: 10.1016/j.ejca.2023.113501] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
The 2022 yearly Think Tank Meeting in Siena, Tuscany (Italy), organized by the Italian Network for Tumor Biotherapy (NIBIT) Foundation, the Parker Institute for Cancer Immunotherapy and the World Immunotherapy Council, included a focus on the future of integrating and expanding the use of targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The conference members exchanged their views on the lessons from targeting CTLA-4 and compared the effect to the impact of blocking Programmed cell death protein 1 (PD1) or its ligand (PDL1). The increasing experience with both therapeutic approaches and their combination suggests that targeting CTLA-4 may lead to more durable responses for a sizeable proportion of patients, though the specific mechanism is not entirely understood. Overcoming toxicity of blocking CTLA-4 is currently being addressed with different doses and dose regimens, especially when combined with PD1/PDL1 blocking antibodies. Novel therapeutics targeting CTLA-4 hold the promise to reduce toxicities and thus allow different combination strategies in the future. On the whole, the consent was that targeting CTLA-4 remains an important strategy to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Maria Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland
| | - Alexander Mm Eggermont
- Princess Máxima Center and the University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands; Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximiliaan University, Munich, Germany
| | - Bernard Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, 4805 NE Glisan St. Suite 2N35 Portland, OR 97213, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, D3500, San Francisco, CA, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson, 1515 Holcombe Blvd, Houston, Texas 77030, USA
| | - James P Allison
- James P Allison Institute, MD Anderson, 1515 Holcombe Blvd, Texas 77030, USA
| | - Michele Maio
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy.
| |
Collapse
|
407
|
Ribeiro JM, Dixon-Douglas J, André F. Moving to ultra-short therapy to cure patients with cancer: a solution for sustainable cancer care. ESMO Open 2024; 9:102238. [PMID: 38350339 PMCID: PMC10875333 DOI: 10.1016/j.esmoop.2024.102238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Affiliation(s)
- J M Ribeiro
- Département de Médecine Oncologique, Gustave Roussy, Villejuif; Gustave Roussy, INSERM U981, PRISM Center, Villejuif, France.
| | - J Dixon-Douglas
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Australia
| | - F André
- Département de Médecine Oncologique, Gustave Roussy, Villejuif; Gustave Roussy, INSERM U981, PRISM Center, Villejuif, France
| |
Collapse
|
408
|
Ellebaek E, Khan S, Bastholt L, Schmidt H, Haslund CA, Donia M, Svane IM. PD-L1 is a biomarker of real-world clinical outcomes for anti-CTLA-4 plus anti-PD-1 or anti-PD-1 monotherapy in metastatic melanoma. Eur J Cancer 2024; 198:113476. [PMID: 38171116 DOI: 10.1016/j.ejca.2023.113476] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Metastatic melanoma (MM) is commonly treated with a combination of nivolumab and ipilimumab, regardless of tumor PD-L1 expression. METHODS We conducted a population-based study including all patients with MM (except ocular melanoma) treated in Denmark with first-line combination therapy or anti-PD-1 monotherapy since January 2017. Baseline data including known prognostic characteristics were used in multivariable and propensity-matched score (PMS) analyses to assess progression-free survival (PFS), melanoma-specific survival (MSS), and overall survival (OS) according to PD-L1 expression. RESULTS We identified 1341 eligible patients, with known PD-L1 status for 1081 patients (43% PD-L1 ≥ 1%, 57% PD-L1 < 1%). PD-L1 ≥ 1% was an independent positive prognostic biomarker for survival in the overall cohort (MSS: HR 0.66, CI 0.52-0.83, p < 0.001). In the PMS PD-L1 ≥ 1% cohort, combination therapy showed similar clinical outcomes to monotherapy (PFS: HR 1.41, CI 0.94-2.11, p = 0.101; MSS: HR 1.21, CI 0.70-2.11, p = 0.49; OS: HR 1.17, CI 0.68-2.00, p = 0.567). In contrast, in the PMS PD-L1 < 1% and in the PMS PD-L1 < 1% BRAF WT cohorts, combination therapy improved PFS (respectively with HR 0.70, CI 0.53-0.93, p = 0.013; and HR 0.54, CI 0.37-0.78, p = 0.001), but did not reach statistically significant improvements of MSS (HR 0.72, CI 0.50-1.02, p = 0.065; and HR 0.79, CI 0.51-1.21, p = 0.278) or OS (HR 0.78, CI 0.56-1.08, p = 0.135; and HR 0.81, CI 0.54-1.21, p = 0.305) compared to monotherapy. CONCLUSION Our findings support previous exploratory analyses of Checkmate-067, highlighting that improved clinical outcomes with combination therapy are not established in unselected patients with high (≥1%) tumor PD-L1 expression.
Collapse
Affiliation(s)
- Eva Ellebaek
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Shawez Khan
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.
| |
Collapse
|
409
|
San-Román-Gil M, Martínez-Delfrade I, Albarrán-Fernández V, Guerrero-Serrano P, Pozas-Pérez J, Chamorro-Pérez J, Rosero-Rodríguez D, Sotoca-Rubio P, Barrill-Corpa AM, Alia-Navarro V, González-Merino C, García-de-Quevedo-Suero C, López V, Ruz-Caracuel I, Perna-Monroy C, Ferreiro-Monteagudo R. Case report: Efficacy of immunotherapy as conversion therapy in dMMR/MSI-H colorectal cancer: a case series and review of the literature. Front Immunol 2024; 15:1352262. [PMID: 38361927 PMCID: PMC10867218 DOI: 10.3389/fimmu.2024.1352262] [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: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Immunotherapy has demonstrated a role in the therapeutic landscape of a small subset of patients with colorectal carcinoma (CRC) that harbor a microsatellite instability (MSI-H) status due to a deficient DNA mismatch repair (dMMR) system. The remarkable responses to immune checkpoint inhibitors (ICIs) are now being tested in the neoadjuvant setting in localized CRC, where the dMMR/MSI-H status can be found in up to 15% of patients, with remarkable results obtained in NICHE2 and 3 trials, among others. This case series aims to report our experience at a tertiary center and provide a comprehensive analysis of the possible questions and challenges to overcome if ICIs were established as standard of care in a neoadjuvant setting, as well as the potential role they may have as conversion therapy not only in locoregional advanced CRC but also in oligometastatic disease.
Collapse
Affiliation(s)
- María San-Román-Gil
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | | | | | - Javier Pozas-Pérez
- Medical Oncology Department, Royal Marsden Hospital, London, United Kingdom
| | - Jesús Chamorro-Pérez
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | - Pilar Sotoca-Rubio
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | - Víctor Alia-Navarro
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | | | - Victoria López
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | | | | | | |
Collapse
|
410
|
Gravina AG, Pellegrino R, Esposito A, Cipullo M, Romeo M, Palladino G, Iodice P, Federico A, Troiani T. The JAK-STAT Pathway as a Therapeutic Strategy in Cancer Patients with Immune Checkpoint Inhibitor-Induced Colitis: A Narrative Review. Cancers (Basel) 2024; 16:611. [PMID: 38339367 PMCID: PMC10854551 DOI: 10.3390/cancers16030611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Immunotherapy has emerged as a pivotal component in the treatment of various malignancies, encompassing lung, skin, gastrointestinal, and head and neck cancers. The foundation of this therapeutic approach lies in immune checkpoint inhibitors (ICI). While ICIs have demonstrated remarkable efficacy in impeding the neoplastic progression of these tumours, their use may give rise to substantial toxicity, notably in the gastrointestinal domain, where ICI colitis constitutes a significant aspect. The optimal positioning of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway inhibitors in the therapeutic management of ICI colitis remains unclear. Numerous reports have highlighted notable improvements in ICI colitis through the application of pan-JAK-STAT inhibitors, with tofacitinib, in particular, reporting evident clinical remission of colitis. The precise mechanism by which JAK-STAT inhibitors may impact the pathogenetic process of ICI colitis remains inadequately understood. However, there is speculation regarding their potential role in modulating memory resident CD8+ T lymphocytes. The elucidation of this mechanism requires further extensive and robust evidence, and ongoing JAK-STAT-based trials are anticipated to contribute valuable insights.
Collapse
Affiliation(s)
- Antonietta Gerarda Gravina
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Raffaele Pellegrino
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Alfonso Esposito
- Oncology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Marina Cipullo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Mario Romeo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Giovanna Palladino
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Patrizia Iodice
- Oncology Division, AORN Ospedali Dei Colli, Monaldi Hospital, Via L. Bianchi, 80131 Naples, Italy
| | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | - Teresa Troiani
- Oncology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| |
Collapse
|
411
|
Farah C, Mignion L, Jordan BF. Metabolic Profiling to Assess Response to Targeted and Immune Therapy in Melanoma. Int J Mol Sci 2024; 25:1725. [PMID: 38339003 PMCID: PMC10855758 DOI: 10.3390/ijms25031725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
There is currently no consensus to determine which advanced melanoma patients will benefit from targeted therapy, immunotherapy, or a combination of both, highlighting the critical need to identify early-response biomarkers to advanced melanoma therapy. The goal of this review is to provide scientific rationale to highlight the potential role of metabolic imaging to assess response to targeted and/or immune therapy in melanoma cancer. For that purpose, a brief overview of current melanoma treatments is provided. Then, current knowledge with respect to melanoma metabolism is described with an emphasis on major crosstalks between melanoma cell metabolism and signaling pathways involved in BRAF-targeted therapy as well as in immune checkpoint inhibition therapies. Finally, preclinical and clinical studies using metabolic imaging and/or profiling to assess response to melanoma treatment are summarized with a particular focus on PET (Positron Emission Tomography) imaging and 13C-MRS (Magnetic Resonance Spectroscopy) methods.
Collapse
Affiliation(s)
- Chantale Farah
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| | - Lionel Mignion
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| | - Bénédicte F. Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| |
Collapse
|
412
|
Yang Y, Wang S, Wang XX, Guo S, Wang H, Shi Q, Tian Y, Wang H, Zhao T, Zhang H, Zhang B, Gao T, Li C, Yi X, Guo W. Tumorous IRE1α facilitates CD8 +T cells-dependent anti-tumor immunity and improves immunotherapy efficacy in melanoma. Cell Commun Signal 2024; 22:83. [PMID: 38291473 PMCID: PMC10826282 DOI: 10.1186/s12964-024-01470-8] [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: 06/24/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Tumor cells frequently suffer from endoplasmic reticulum (ER) stress. Previous studies have extensively elucidated the role of tumorous unfolded protein response in melanoma cells, whereas the effect on tumor immunology and the underlying mechanism remain elusive. METHODS Bioinformatics, biochemical assays and pre-clinical mice model were employed to demonstrate the role of tumorous inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in anti-tumor immunity and the underlying mechanism. RESULTS We firstly found that IRE1α signaling activation was positively associated with the feature of tumor-infiltrating lymphocytes. Then, pharmacological ER stress induction by HA15 exerted prominent anti-tumor effect in immunocompetent mice and was highly dependent on CD8+T cells, paralleled with the reshape of immune cells in tumor microenvironment via tumorous IRE1α-XBP1 signal. Subsequently, tumorous IRE1α facilitated the expression and secretion of multiple chemokines and cytokines via XBP1-NF-κB axis, leading to increased infiltration and anti-tumor capacity of CD8+T cells. Ultimately, pharmacological induction of tumorous ER stress by HA15 brought potentiated therapeutic effect along with anti-PD-1 antibody on melanoma in vivo. CONCLUSIONS Tumorous IRE1α facilitates CD8+T cells-dependent anti-tumor immunity and improves immunotherapy efficacy by regulating chemokines and cytokines via XBP1-NF-κB axis. The combination of ER stress inducer and anti-PD-1 antibody could be promising for increasing the efficacy of melanoma immunotherapy.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sijia Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiang-Xu Wang
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qiong Shi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangzi Tian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hao Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tao Zhao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hengxiang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Baolu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
413
|
Goldstein DA, Saltz LB, Pond GR, Tannock IF. Pharmacological class effects of anticancer drugs: opportunities for decreasing healthcare spending. BMJ ONCOLOGY 2024; 3:e000287. [PMID: 39886138 PMCID: PMC11234998 DOI: 10.1136/bmjonc-2023-000287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2025]
Abstract
In the field of general medicine, class effects, or therapeutic interchangeability, have been declared for several families of drugs including statins, calcium channel blockers and ACE inhibitors. The existence of such class effects enables healthcare payers to negotiate for substantially lower drug prices, thereby reducing financial toxicity, both at an individual and societal levels. Until now, the existence of class effects in oncology has been considered rare. Here, we review evidence from clinical trials that supports the existence of class effects for several types of anticancer drugs. These class effects in oncology should be exploited to reduce healthcare costs.
Collapse
Affiliation(s)
- Daniel A Goldstein
- Medical Oncology, Rabin Medical Center Davidoff Cancer Center, Petah Tikva, Central, Israel
| | - Leonard B Saltz
- Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gregory R Pond
- Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Ian F Tannock
- Medical Oncology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
414
|
Keshari S, Shavkunov AS, Miao Q, Saha A, Williams CD, Highsmith AM, Pineda JE, Alspach E, Hu KH, Pauken KE, Chen K, Gubin MM. Neoantigen Cancer Vaccines and Different Immune Checkpoint Therapies Each Utilize Both Converging and Distinct Mechanisms that in Combination Enable Synergistic Therapeutic Efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.20.570816. [PMID: 38187708 PMCID: PMC10769249 DOI: 10.1101/2023.12.20.570816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The goal of therapeutic cancer vaccines and immune checkpoint therapy (ICT) is to eliminate cancer by expanding and/or sustaining T cells with anti-tumor capabilities. However, whether cancer vaccines and ICT enhance anti-tumor immunity by distinct or overlapping mechanisms remains unclear. Here, we compared effective therapeutic tumor-specific mutant neoantigen (NeoAg) cancer vaccines with anti-CTLA-4 and/or anti-PD-1 ICT in preclinical models. Both NeoAg vaccines and ICT induce expansion of intratumoral NeoAg-specific CD8 T cells, though the degree of expansion and acquisition of effector activity was much more substantial following NeoAg vaccination. Further, we found that NeoAg vaccines are particularly adept at inducing proliferating and stem-like NeoAg-specific CD8 T cells. Single cell T cell receptor (TCR) sequencing revealed that TCR clonotype expansion and diversity of NeoAg-specific CD8 T cells relates to their phenotype and functional state associated with specific immunotherapies employed. Effective NeoAg vaccines and ICT required both CD8 and CD4 T cells. While NeoAg vaccines and anti-PD-1 affected the CD4 T cell compartment, it was to less of an extent than observed with anti-CTLA-4, which notably induced ICOS+Bhlhe40+ Th1-like CD4 T cells and, when combined with anti-PD-1, a small subset of Th2-like CD4 T cells. Although effective NeoAg vaccines or ICT expanded intratumoral M1-like iNOS+ macrophages, NeoAg vaccines expanded rather than suppressed (as observed with ICT) M2-like CX3CR1+CD206+ macrophages, associated with the vaccine adjuvant. Further, combining NeoAg vaccination with ICT induced superior efficacy compared to either therapy in isolation, highlighting the utility of combining these modalities to eliminate cancer.
Collapse
Affiliation(s)
- Sunita Keshari
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander S. Shavkunov
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akata Saha
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charmelle D. Williams
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna M. Highsmith
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Josué E. Pineda
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elise Alspach
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Kenneth H. Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristen E. Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew M. Gubin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
415
|
Santry LA, van Vloten JP, AuYeung AWK, Mould RC, Yates JGE, McAusland TM, Petrik JJ, Major PP, Bridle BW, Wootton SK. Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer. Front Microbiol 2024; 15:1325558. [PMID: 38328418 PMCID: PMC10847535 DOI: 10.3389/fmicb.2024.1325558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024] Open
Abstract
Introduction Tumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and regulatory T cells (Tregs). Methods To reverse this immunosuppression, we engineered mesogenic Newcastle disease virus (NDV) to express immunological checkpoint inhibitors anti-cytotoxic T lymphocyte antigen-4 and soluble programmed death protein-1. Results Intratumoral administration of recombinant NDV (rNDV) to mice bearing intradermal B16-F10 melanomas or subcutaneous CT26LacZ colon carcinomas led to significant changes in the tumor-infiltrating lymphocyte profiles. Vectorizing immunological checkpoint inhibitors in NDV increased activation of intratumoral natural killer cells and cytotoxic T cells and decreased Tregs and MDSCs, suggesting induction of a pro-inflammatory state with greater infiltration of activated CD8+ T cells. These notable changes translated to higher ratios of activated effector/suppressor tumor-infiltrating lymphocytes in both cancer models, which is a promising prognostic marker. Whereas all rNDV-treated groups showed evidence of tumor regression and increased survival in the CT26LacZ and B16-F10, only treatment with NDV expressing immunological checkpoint blockades led to complete responses compared to tumors treated with NDV only. Discussion These data demonstrated that NDV expressing immunological checkpoint inhibitors could reverse the immunosuppressive state of tumor microenvironments and enhance tumor-specific T cell responses.
Collapse
Affiliation(s)
- Lisa A. Santry
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob P. van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Amanda W. K. AuYeung
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Robert C. Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob G. E. Yates
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Thomas M. McAusland
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - James J. Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
416
|
Meraz-Torres F, Niessner H, Plöger S, Riel S, Schörg B, Casadei N, Kneilling M, Schaller M, Flatz L, Macek B, Eigentler T, Rieß O, Garbe C, Amaral T, Sinnberg T. Augmenting MEK inhibitor efficacy in BRAF wild-type melanoma: synergistic effects of disulfiram combination therapy. J Exp Clin Cancer Res 2024; 43:30. [PMID: 38263136 PMCID: PMC10804659 DOI: 10.1186/s13046-023-02941-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND MEK inhibitors (MEKi) were shown to be clinically insufficiently effective in patients suffering from BRAF wild-type (BRAF WT) melanoma, even if the MAPK pathway was constitutively activated due to mutations in NRAS or NF-1. Thus, novel combinations are needed to increase the efficacy and duration of response to MEKi in BRAF WT melanoma. Disulfiram and its metabolite diethyldithiocarbamate are known to have antitumor effects related to cellular stress, and induction of endoplasmic reticulum (ER) stress was found to synergize with MEK inhibitors in NRAS-mutated melanoma cells. Therefore, we investigated the combination of both therapeutics to test their effects on BRAF-WT melanoma cells and compared them with monotherapy using the MEKi trametinib. METHODS The effects of combined therapy with disulfiram or its metabolite diethyldithiocarbamate and the MEKi trametinib were evaluated in a series of BRAF-WT melanoma cell lines by measuring cell viability and apoptosis induction. Cytotoxicity was additionally assessed in 3D spheroids, ex vivo melanoma slice cultures, and in vivo xenograft mouse models. The response of melanoma cells to treatment was studied at the RNA and protein levels to decipher the mode of action. Intracellular and intratumoral copper measurements were performed to investigate the role of copper ions in the antitumor cytotoxicity of disulfiram and its combination with the MEKi. RESULTS Diethyldithiocarbamate enhanced trametinib-induced cytotoxicity and apoptosis induction in 2D and 3D melanoma culture models. Mechanistically, copper-dependent induction of oxidative stress and ER stress led to Janus kinase (JNK)-mediated apoptosis in melanoma cells. This mechanism was also detectable in patient-derived xenograft melanoma models and resulted in a significantly improved therapeutic effect compared to monotherapy with the MEKi trametinib. CONCLUSIONS Disulfiram and its metabolite represent an attractive pharmaceutical approach to induce ER stress in melanoma cells that potentiates the antitumor effect of MEK inhibition and may be an interesting candidate for combination therapy of BRAF WT melanoma.
Collapse
Affiliation(s)
| | - Heike Niessner
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
| | - Sarah Plöger
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Simon Riel
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Barbara Schörg
- Department of Preclinical Imaging and Radiopharmacy, Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, University of Tübingen, Tübingen, 72076, Germany
| | - Nicolas Casadei
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Manfred Kneilling
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
- Department of Preclinical Imaging and Radiopharmacy, Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, University of Tübingen, Tübingen, 72076, Germany
| | - Martin Schaller
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Olaf Rieß
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
| | - Tobias Sinnberg
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany.
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
417
|
Reitmajer M, Leiter U, Nanz L, Amaral T, Flatz L, Garbe C, Forschner A. Long-term survival of stage IV melanoma patients: evaluation on 640 melanoma patients entering stage IV between 2014 and 2017. J Cancer Res Clin Oncol 2024; 150:15. [PMID: 38238578 PMCID: PMC10796594 DOI: 10.1007/s00432-023-05533-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 01/22/2024]
Abstract
PURPOSE Since the introduction of immune checkpoint inhibitors (ICI) and targeted therapies (TT), survival rates of metastatic melanoma patients have increased significantly and complete remissions are no longer rarities. Consequently, there is an increasing number of long-term survivors who have not yet been comprehensively characterized. METHODS We included melanoma patients who entered stage IV between 2014 and 2017 and survived at least 5 years after entering stage IV. Descriptive statistics were performed to characterize the applied systemic therapies, response rates and to report which of these patients are still alive today. RESULTS 640 patients entered stage IV at the University Hospital Tuebingen. Of these, 207 patients (32%) were still alive at least 5 years after entering stage IV. Details of applied therapies and response rates were available in 176 patients (85%). About 90% of patients (n = 159) were still alive at the time of analysis. Median survival since first stage IV diagnosis was 6.0 years (range 5-9 years). An impressive majority of patients (n = 146, 83%) were no longer receiving systemic therapy at the time of evaluation. Complete remission under first line systemic therapy was seen in 36% of the patients. CONCLUSION This dataset comprises the largest available cohort of long-term surviving stage IV melanoma patients. Since 90% of patients in our cohort are still alive today, we expect an increasing number of long-term survivors in the future. Our data indicate the need for specific follow-up programs addressing the needs of long-term survivors.
Collapse
Affiliation(s)
- Markus Reitmajer
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany.
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| | - Lena Nanz
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, 72076, Tuebingen, Germany
| |
Collapse
|
418
|
Chen W, He Y, Zhou G, Chen X, Ye Y, Zhang G, Liu H. Multiomics characterization of pyroptosis in the tumor microenvironment and therapeutic relevance in metastatic melanoma. BMC Med 2024; 22:24. [PMID: 38229080 PMCID: PMC10792919 DOI: 10.1186/s12916-023-03175-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 11/14/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Pyroptosis, mediated by gasdermins with the release of multiple inflammatory cytokines, has emerged as playing an important role in targeted therapy and immunotherapy due to its effectiveness at inhibiting tumor growth. Melanoma is one of the most commonly used models for immunotherapy development, though an inadequate immune response can occur. Moreover, the development of pyroptosis-related therapy and combinations with other therapeutic strategies is limited due to insufficient understanding of the role of pyroptosis in the context of different tumor immune microenvironments (TMEs). METHODS Here, we present a computational model (pyroptosis-related gene score, PScore) to assess the pyroptosis status. We applied PScore to 1388 melanoma samples in our in-house cohort and eight other publicly available independent cohorts and then calculated its prognostic power of and potential as a predictive marker of immunotherapy efficacy. Furthermore, we performed association analysis for PScore and the characteristics of the TME by using bulk, single-cell, and spatial transcriptomics and assessed the association of PScore with mutation status, which contributes to targeted therapy. RESULTS Pyroptosis-related genes (PRGs) showed distinct expression patterns and prognostic predictive ability in melanoma. Most PRGs were associated with better survival in metastatic melanoma. Our PScore model based on genes associated with prognosis exhibits robust performance in survival prediction in multiple metastatic melanoma cohorts. We also found PScore to be associated with BRAF mutation and correlate positively with multiple molecular signatures, such as KRAS signaling and the IFN gamma response pathway. Based on our data, melanoma with an immune-enriched TME had a higher PScore than melanoma with an immune-depleted or fibrotic TME. Additionally, monocytes had the highest PScore and malignant cells and fibroblasts the lowest PScore based on single-cell and spatial transcriptome analyses. Finally, a higher PScore was associated with better therapeutic efficacy of immune checkpoint blockade, suggesting the potential of pyroptosis to serve as a marker of immunotherapy response. CONCLUSIONS Collectively, our findings indicate that pyroptosis is a prognostic factor and is associated with the immune response in metastatic melanoma, as based on multiomics data. Our results provide a theoretical basis for drug combination and reveal potential immunotherapy response markers.
Collapse
Affiliation(s)
- Wenqiong Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Yi He
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Guowei Zhou
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Youqiong Ye
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Guanxiong Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Hong Liu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.
- Big Data Institute, Central South University, Changsha, 410083, China.
| |
Collapse
|
419
|
Wang T, Guo K, Yang S, Zhang D, Cui H, Yin J, Yuan S, Wang Y, Qi Y, Wu W. Identification and validation of SLCO4C1 as a biological marker in hepatocellular carcinoma based on anoikis classification features. Aging (Albany NY) 2024; 16:1440-1462. [PMID: 38226966 PMCID: PMC10866452 DOI: 10.18632/aging.205438] [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: 09/18/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) exhibits a high degree of invasiveness and is closely associated with rapid disease progression. Multiple lines of evidence indicate a strong correlation between anoikis resistance and tumor progression, invasion, and metastasis. Nevertheless, the classification of anoikis in HCC and the investigation of novel biological target mechanisms in this context continue to pose challenges, requiring further exploration. METHODS Combined with HCC samples from TCGA, GEO and ICGC databases, cluster analysis was conducted on anoikis genes, revealing novel patterns among different subtypes. Significant gene analysis of different gene subtypes was performed using WCGNA. The anoikis prognostic risk model was established by Lasso-Cox. Go, KEGG, and GSEA were applied to investigate pathway enrichment primarily observed in risk groups. We compared the disparities in immune infiltration, TMB, tumor microenvironment (TME), and drug sensitivity between the two risk groups. RT-qPCR and Western blotting were performed to validate the expression levels of SLCO4C1 in HCC. The biological functions of SLCO4C1 in HCC cells were assessed through various experiments, including CCK8 assay, colony formation assay, invasion migration assay, wound healing assay, and flow cytometry analysis. RESULTS HCC was divided into 2 anoikis subtypes, and the subtypeB had a better prognosis. An anoikis prognostic model based on 12 (COPZ2, ACTG2, IFI27, SPP1, EPO, SLCO4C1, RAB26, STC2, RAC3, NQO1, MYCN, HSPA1B) risk genes is important for survival and prognosis. Significant differences were observed in immune cell infiltration, TME, and drug sensitivity analysis between the risk groups. SLCO4C1 was downregulated in HCC. SLCO4C1 downregulation promoted the proliferation, invasion, migration, and apoptosis of HCC cells. The tumor-suppressive role of SLCO4C1 in HCC has been confirmed. CONCLUSIONS Our study presents a novel anoikis classification method for HCC that reveals the association between anoikis features and HCC. The anoikis feature is a critical biomarker bridging tumor cell death and tumor immunity. In this study, we provided the first evidence of SLCO4C1 functioning as a tumor suppressor in HCC.
Collapse
Affiliation(s)
- Tianbing Wang
- Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
| | - Kai Guo
- Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
| | - Shoushan Yang
- Anhui No. 2 Provincial People’s Hospital Clinical College of Anhui Medical University, Hefei 230000, China
- Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
- Department of General Surgery, Luan Fourth People’s Hospital, Luan 237000, China
| | - Di Zhang
- Clinical Genomic Center, Hefei KingMed for Clinical Laboratory, Hefei 230000, China
| | - Haodong Cui
- Anhui No. 2 Provincial People’s Hospital Clinical College of Anhui Medical University, Hefei 230000, China
- Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Jimin Yin
- Anhui No. 2 Provincial People’s Hospital Clinical College of Anhui Medical University, Hefei 230000, China
- Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| | - Shuhui Yuan
- Anhui Huaheng Biotechnology Co., Ltd., Hefei 230000, China
| | - Yong Wang
- Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
| | - Yong Qi
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| | - Wenyong Wu
- Anhui No. 2 Provincial People’s Hospital Clinical College of Anhui Medical University, Hefei 230000, China
- Anhui No. 2 Provincial People’s Hospital, Hefei 230000, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, China
| |
Collapse
|
420
|
Losurdo A, Dipasquale A, Giordano L, Persico P, Lorenzi E, Di Muzio A, Barigazzi C, Korolewicz J, Mehan A, Mohammed O, Scheiner B, Pinato DJ, Santoro A, Simonelli M. Refining patient selection for next-generation immunotherapeutic early-phase clinical trials with a novel and externally validated prognostic nomogram. Front Immunol 2024; 15:1323151. [PMID: 38298193 PMCID: PMC10828843 DOI: 10.3389/fimmu.2024.1323151] [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: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Introduction Identifying which patient may benefit from immunotherapeutic early-phase clinical trials is an unmet need in drug development. Among several proposed prognostic scores, none has been validated in patients receiving immunomodulating agents (IMAs)-based combinations. Patients and methods We retrospectively collected data of 208 patients enrolled in early-phase clinical trials investigating IMAs at our Institution, correlating clinical and blood-based variables with overall survival (OS). A retrospective cohort of 50 patients treated with IMAs at Imperial College (Hammersmith Hospital, London, UK) was used for validation. Results A total of 173 subjects were selected for analyses. Most frequent cancers included non-small cell lung cancer (26%), hepatocellular carcinoma (21.5%) and glioblastoma (13%). Multivariate analysis (MVA) revealed 3 factors to be independently associated with OS: line of treatment (second and third vs subsequent, HR 0.61, 95% CI 0.40-0.93, p 0.02), serum albumin as continuous variable (HR 0.57, 95% CI 0.36-0.91, p 0.02) and number of metastatic sites (<3 vs ≥3, HR 0.68, 95% CI 0.48-0.98, p 0.04). After splitting albumin value at the median (3.84 g/dL), a score system was capable of stratifying patients in 3 groups with significantly different OS (p<0.0001). Relationship with OS reproduced in the external cohort (p=0.008). Then, from these factors we built a nomogram. Conclusions Prior treatment, serum albumin and number of metastatic sites are readily available prognostic traits in patients with advanced malignancies participating into immunotherapy early-phase trials. Combination of these factors can optimize patient selection at study enrollment, maximizing therapeutic intent.
Collapse
Affiliation(s)
- Agnese Losurdo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Laura Giordano
- Biostatistic Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Elena Lorenzi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Antonio Di Muzio
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Chiara Barigazzi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - James Korolewicz
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Aman Mehan
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Oreoluwa Mohammed
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Benhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - David J. Pinato
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Division of Oncology, Department of Translational Medicine (DIMET), Università del Piemonte Orientale A. Avogadro, Novara, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| |
Collapse
|
421
|
Curkovic NB, Bai K, Ye F, Johnson DB. Incidence of Cutaneous Immune-Related Adverse Events and Outcomes in Immune Checkpoint Inhibitor-Containing Regimens: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:340. [PMID: 38254829 PMCID: PMC10814132 DOI: 10.3390/cancers16020340] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are used to treat many cancers, and cutaneous immune-related adverse events (cirAEs) are among the most frequently encountered toxic effects. Understanding the incidence and prognostic associations of cirAEs is of importance as their uses in different settings, combinations, and tumor types expand. To evaluate the incidence of cirAEs and their association with outcome measures across a variety of ICI regimens and cancers, we performed a systematic review and meta-analysis of published trials of anti-programmed death-1/ligand-1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) ICIs, both alone and in combination with chemotherapy, antiangiogenic agents, or other ICIs in patients with melanoma, renal cell carcinoma, non-small cell lung cancer, and urothelial carcinoma. Key findings of our study include variable cirAE incidence among tumors and ICI regimens, positive association with increased cirAE incidence and response rate, as well as significant association between increased vitiligo incidence and overall survival. Across 174 studies, rash, pruritis, and vitiligo were the most reported cirAEs, with incidences of 16.7%, 18.0%, and 6.6%, respectively. Higher incidence of cirAEs was associated with ICI combination regimens and with CTLA-4-containing regimens, particularly with higher doses of ipilimumab, as compared to PD-1/L1 monotherapies. Outcome measures including response rate and progression-free survival were positively correlated with incidence of cirAEs. The response rate and incidence of pruritis, vitiligo, and rash were associated with expected rises in incidence of 0.17% (p = 0.0238), 0.40% (p = 0.0010), and 0.18% (p = 0.0413), respectively. Overall survival was positively correlated with the incidence of pruritis, vitiligo, and rash; this association was significant for vitiligo (p = 0.0483). Our analysis provides benchmark incidence rates for cirAEs and links cirAEs with favorable treatment outcomes at a study level across diverse solid tumors and multiple ICI regimens.
Collapse
Affiliation(s)
- Nina B. Curkovic
- School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Kun Bai
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Fei Ye
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Douglas B. Johnson
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
422
|
Shen H, Mullen L, Ojo OA, Xing C, Yassin A, Lewis Z, Bonner JA, Shi LZ. HIF1α-glycolysis engages activation-induced cell death to drive IFN-γ induction in hypoxic T cells. RESEARCH SQUARE 2024:rs.3.rs-3830704. [PMID: 38260594 PMCID: PMC10802708 DOI: 10.21203/rs.3.rs-3830704/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The role of HIF1α-glycolysis in regulating IFN-γ induction in hypoxic T cells is unknown. Given that hypoxia is a common feature in a wide array of pathophysiological contexts such as tumor and that IFN-γ is instrumental for protective immunity, it is of great significance to gain a clear idea on this. Combining pharmacological and genetic gain-of-function and loss-of-function approaches, we find that HIF1α-glycolysis controls IFN-γ induction in both human and mouse T cells activated under hypoxia. Specific deletion of HIF1α in T cells (HIF1α-/-) and glycolytic inhibition significantly abrogate IFN-γ induction. Conversely, HIF1α stabilization in T cells by hypoxia and VHL deletion (VHL-/-) promotes IFN-γ production. Mechanistically, reduced IFN-γ production in hypoxic HIF1α-/- T cells is due to attenuated activation-induced cell death but not proliferative defect. We further show that depletion of intracellular acetyl-CoA is a key metabolic underlying mechanism. Hypoxic HIF1α-/- T cells are less able to kill tumor cells, and HIF1α-/- tumor-bearing mice are not responsive to immune checkpoint blockade (ICB) therapy, indicating loss of HIF1α in T cells is a major mechanism of therapeutic resistance to ICBs. Importantly, acetate supplementation restores IFN-γ production in hypoxic HIF1α-/- T cells and re-sensitizes HIF1α-/- tumor-bearing mice to ICBs, providing an effective strategy to overcome ICB resistance. Taken together, our results highlight T cell HIF1α-anaerobic glycolysis as a principal mediator of IFN-γ induction and anti-tumor immunity. Considering that acetate supplementation (i.e., glycerol triacetate (GTA)) is approved to treat infants with Canavan disease, we envision a rapid translation of our findings, justifying further testing of GTA as a repurposed medicine for ICB resistance, a pressing unmet medical need.
Collapse
Affiliation(s)
- Hongxing Shen
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Logan Mullen
- Genomics Core Laboratory, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, 99775, USA
| | - Oluwagbemiga A. Ojo
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Chuan Xing
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Abdelrahman Yassin
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Zach Lewis
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - James A. Bonner
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, UAB-SOM, USA
| | - Lewis Zhichang Shi
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, UAB-SOM, USA
- Department of Microbiology and Immunology Institute, UAB-SOM, USA
- Department of Pharmacology and Toxicology, UAB-SOM, USA
- Immunology Institute, UAB-SOM, USA
| |
Collapse
|
423
|
Su J, Fu Y, Cui Z, Abidin Z, Yuan J, Zhang X, Li R, Zhao C. Relatlimab: a novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front Pharmacol 2024; 14:1349081. [PMID: 38269271 PMCID: PMC10806167 DOI: 10.3389/fphar.2023.1349081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Relatlimab is a type of human immunoglobulin G4 monoclonal blocking antibody. It is the world's first Lymphocyte-Activation Gene-3 (LAG-3) inhibitor and the third immune checkpoint inhibitor with clinical application, following PD-1 and CTLA-4. Relatlimab can bind to the LAG-3 receptor which blocks the interaction between LAG-3 and its ligand to reduce LAG-3 pathway-mediated immunosuppression and promote T-cell proliferation, inducing tumor cell death. On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab developed by Bristol Myers Squibb with nivolumab, under the brand name Opdualag for the treatment of unresectable or metastatic melanoma in adult and pediatric patients aged 12 and older. This study comprehensively describes the mechanism of action and clinical trials of relatlimab and a brief overview of immune checkpoint drugs currently used for the treatment of melanoma.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Yiting Fu
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zitong Cui
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Zain Abidin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jingsong Yuan
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Xinmiao Zhang
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Runmin Li
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
424
|
Peisen F, Gerken A, Dahm I, Nikolaou K, Eigentler T, Amaral T, Moltz JH, Othman AE, Gatidis S. Pre-treatment 18F-FDG-PET/CT parameters as biomarkers for progression free survival, best overall response and overall survival in metastatic melanoma patients undergoing first-line immunotherapy. PLoS One 2024; 19:e0296253. [PMID: 38180971 PMCID: PMC10769042 DOI: 10.1371/journal.pone.0296253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Checkpoint inhibitors have drastically improved the therapy of patients with advanced melanoma. 18F-FDG-PET/CT parameters might act as biomarkers for response and survival and thus can identify patients that do not benefit from immunotherapy. However, little literature exists on the association of baseline 18F-FDG-PET/CT parameters with progression free survival (PFS), best overall response (BOR), and overall survival (OS). MATERIALS AND METHODS Using a whole tumor volume segmentation approach, we investigated in a retrospective registry study (n = 50) whether pre-treatment 18F-FDG-PET/CT parameters of three subgroups (tumor burden, tumor glucose uptake and non-tumoral hematopoietic tissue metabolism), can act as biomarkers for the primary endpoints PFS and BOR as well as for the secondary endpoint OS. RESULTS Compared to the sole use of clinical parameters, baseline 18F-FDG-PET/CT parameters did not significantly improve a Cox proportional-hazard model for PFS (C-index/AIC: 0.70/225.17 and 0.68/223.54, respectively; p = 0.14). A binomial logistic regression analysis for BOR was not statistically significant (χ2(15) = 16.44, p = 0.35), with a low amount of explained variance (Nagelkerke's R2 = 0.38). Mean FDG uptake of the spleen contributed significantly to a Cox proportional-hazard model for OS (HR 3.55, p = 0.04). CONCLUSIONS The present study could not confirm the capability of the pre-treatment 18F-FDG-PET/CT parameters tumor burden, tumor glucose uptake and non-tumoral hematopoietic tissue metabolism to act as biomarkers for PFS and BOR in metastatic melanoma patients receiving first-line immunotherapy. The documented potential of 18F-FDG uptake by immune-mediating tissues such as the spleen to act as a biomarker for OS has been reproduced.
Collapse
Affiliation(s)
- Felix Peisen
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Isabel Dahm
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Image-guided and Functionally Instructed Tumor Therapies (iFIT), The Cluster of Excellence (EXC 2180), Tuebingen, Germany
| | - Thomas Eigentler
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humbolt-Universität zu Berlin, Berlin, Germany
| | - Teresa Amaral
- Center of Dermato-Oncology, Department of Dermatology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Ahmed E. Othman
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Institute of Neuroradiology, Johannes Gutenberg University Hospital Mainz, Mainz, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
- Max Planck Institute for Intelligent Systems, Tuebingen, Germany
| |
Collapse
|
425
|
Weiss SA, Sznol M, Shaheen M, Berciano-Guerrero MÁ, Couselo EM, Rodríguez-Abreu D, Boni V, Schuchter LM, Gonzalez-Cao M, Arance A, Wei W, Ganti AK, Hauke RJ, Berrocal A, Iannotti NO, Hsu FJ, Kluger HM. A Phase II Trial of the CD40 Agonistic Antibody Sotigalimab (APX005M) in Combination with Nivolumab in Subjects with Metastatic Melanoma with Confirmed Disease Progression on Anti-PD-1 Therapy. Clin Cancer Res 2024; 30:74-81. [PMID: 37535056 PMCID: PMC10767304 DOI: 10.1158/1078-0432.ccr-23-0475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
PURPOSE Disease progression during or after anti-PD-1-based treatment is common in advanced melanoma. Sotigalimab is a CD40 agonist antibody with a unique epitope specificity and Fc receptor binding profile optimized for activation of CD40-expressing antigen-presenting cells. Preclinical data indicated that CD40 agonists combined with anti-PD1 could overcome resistance to anti-PD-1. PATIENTS AND METHODS We conducted a multicenter, open-label, phase II trial to evaluate the combination of sotigalimab 0.3 mg/kg and nivolumab 360 mg every 3 weeks in patients with advanced melanoma following confirmed disease progression on a PD-1 inhibitor. The primary objective was to determine the objective response rate (ORR). RESULTS Thirty-eight subjects were enrolled and evaluable for safety. Thirty-three were evaluable for activity. Five confirmed partial responses (PR) were observed for an ORR of 15%. Two PRs are ongoing at 45.9+ and 26+ months, whereas the other three responders relapsed at 41.1, 18.7, and 18.4 months. The median duration of response was at least 26 months. Two additional patients had stable disease for >6 months. Thirty-four patients (89%) experienced at least one adverse event (AE), and 13% experienced a grade 3 AE related to sotigalimab. The most common AEs were pyrexia, chills, nausea, fatigue, pruritus, elevated liver function, rash, vomiting, headache, arthralgia, asthenia, myalgia, and diarrhea. There were no treatment-related SAEs, deaths, or discontinuation of sotigalimab due to AEs. CONCLUSIONS Sotigalimab plus nivolumab had a favorable safety profile consistent with the toxicity profiles of each agent. The combination resulted in durable and prolonged responses in a subset of patients with anti-PD-1-resistant melanoma, warranting further evaluation in this setting. See related commentary by Wu and Luke, p. 9.
Collapse
Affiliation(s)
- Sarah A. Weiss
- Yale University School of Medicine, New Haven, Connecticut
| | - Mario Sznol
- Yale University School of Medicine, New Haven, Connecticut
| | | | - Miguel-Ángel Berciano-Guerrero
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | | | | | - Valentina Boni
- START Madrid-CIOCC, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Lynn M. Schuchter
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Gonzalez-Cao
- Instituto Oncológico, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Ana Arance
- Hospital Clínic Barcelona, Barcelona, Spain
| | - Wei Wei
- Yale University School of Medicine, New Haven, Connecticut
| | - Apar Kishor Ganti
- VA Nebraska Western Iowa Healthcare System and University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | | | | | |
Collapse
|
426
|
Lin H, Xu Y, Lin C. Heterogeneity and subtypes of CD4 + regulatory T cells: implications for tumor therapy. Front Immunol 2024; 14:1291796. [PMID: 38250084 PMCID: PMC10796559 DOI: 10.3389/fimmu.2023.1291796] [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: 09/10/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
In the conventional view, CD4+ regulatory T cell (Treg) represents a subset of lymphocytes that involve the perception and negative regulation of the immune response. CD4+Treg plays an important role in the maintenance of immune homeostasis and immune tolerance. However, recent studies have revealed that CD4+Treg do not suppress the immune response in some diseases, but promote inflammatory injury or inhibit tissue remodeling, suggesting the functional heterogeneity of CD4+Treg. Their involvement in tumor pathogenesis is more complex than previously understood. This article reviews the relevant research on the heterogeneity of CD4+Treg, subtype classification, and their relationship with tumor therapy.
Collapse
Affiliation(s)
- Hanqing Lin
- Department of Otolaryngology, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Fujian Medical University, Fuzhou, China
| | - Yuanteng Xu
- Department of Otolaryngology, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Fujian Medical University, Fuzhou, China
| | - Chang Lin
- Department of Otolaryngology, Fujian Institute of Otorhinolaryngology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Fujian Medical University, Fuzhou, China
| |
Collapse
|
427
|
Fiorentino V, Pizzimenti C, Franchina M, Pepe L, Russotto F, Tralongo P, Micali MG, Militi GB, Lentini M. Programmed Cell Death Ligand 1 Immunohistochemical Expression and Cutaneous Melanoma: A Controversial Relationship. Int J Mol Sci 2024; 25:676. [PMID: 38203846 PMCID: PMC10779806 DOI: 10.3390/ijms25010676] [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: 10/09/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Cutaneous melanoma (CM) is traditionally considered one of the most "immunogenic" tumors, eliciting a high immune response. However, despite the presence of tumor-infiltrating lymphocytes (TILs), melanoma cells use strategies to suppress antitumor immunity and avoid being eliminated by immune surveillance. The PD-1 (programmed death-1)/PD-L1 (programmed death-ligand 1) axis is a well-known immune escape system adopted by neoplastic cells. Therefore, immunotherapy with PD-1 and PD-L1 inhibitors is quickly becoming the main treatment approach for metastatic melanoma patients. However, the clinical utility of PD-L1 expression assessment in CM is controversial, and the interpretation of PD-L1 scores in clinical practice is still a matter of debate. Nonetheless, the recent literature data show that by adopting specific PD-L1 assessment methods in melanoma samples, a correlation between the expression of such a biomarker and a positive response to PD-1-based immunotherapy can be seen. Our review aims to describe the state-of-the-art knowledge regarding the prognostic and predictive role of PD-L1 expression in CM while also referring to possible biological explanations for the variability in its expressions and related treatment responses.
Collapse
Affiliation(s)
- Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Ludovica Pepe
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Fernanda Russotto
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Pietro Tralongo
- Department of Women, Children and Public Health Sciences, Catholic University of the Sacred Heart, Agostino Gemelli IRCCS University Hospital Foundation, 00168 Rome, Italy;
| | - Marina Gloria Micali
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| | - Gaetano Basilio Militi
- Department of Sciences for Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, 90133 Palermo, Italy;
| | - Maria Lentini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (M.F.); (L.P.); (F.R.); (M.G.M.); (M.L.)
| |
Collapse
|
428
|
Collier C, Wucherer K, McWhorter M, Jenkins C, Bartlett A, Roychoudhuri R, Eil R. Intracellular K+ Limits T-cell Exhaustion and Preserves Antitumor Function. Cancer Immunol Res 2024; 12:36-47. [PMID: 38063845 PMCID: PMC10765769 DOI: 10.1158/2326-6066.cir-23-0319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/08/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
T cells are often compromised within cancers, allowing disease progression. We previously found that intratumoral elevations in extracellular K+, related to ongoing cell death, constrained CD8+ T-cell Akt-mTOR signaling and effector function. To alleviate K+-mediated T-cell dysfunction, we pursued genetic means to lower intracellular K+. CD8+ T cells robustly and dynamically express the Na+/K+ ATPase, among other K+ transporters. CRISPR-Cas9-mediated disruption of the Atp1a1 locus lowered intracellular K+ and elevated the resting membrane potential (i.e., Vm, Ψ). Despite compromised Ca2+ influx, Atp1a1-deficient T cells harbored tonic hyperactivity in multiple signal transduction cascades, along with a phenotype of exhaustion in mouse and human CD8+ T cells. Provision of exogenous K+ restored intracellular levels in Atp1a1-deficient T cells and prevented damaging levels of reactive oxygen species (ROS), and both antioxidant treatment and exogenous K+ prevented Atp1a1-deficient T-cell exhaustion in vitro. T cells lacking Atp1a1 had compromised persistence and antitumor activity in a syngeneic model of orthotopic murine melanoma. Translational application of these findings will require balancing the beneficial aspects of intracellular K+ with the ROS-dependent nature of T-cell effector function. See related Spotlight by Banuelos and Borges da Silva, p. 6.
Collapse
Affiliation(s)
- Camille Collier
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Kelly Wucherer
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Matthew McWhorter
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Chelsea Jenkins
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Alexandra Bartlett
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Robert Eil
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
429
|
Jiang Y, Chen H, Lin T, Zhang C, Shen J, Chen J, Zhao Y, Xu W, Wang G, Huang P. Ultrasound-activated prodrug-loaded liposome for efficient cancer targeting therapy without chemotherapy-induced side effects. J Nanobiotechnology 2024; 22:2. [PMID: 38169390 PMCID: PMC10763105 DOI: 10.1186/s12951-023-02195-5] [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: 09/12/2023] [Accepted: 11/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Off-targeted distribution of chemotherapeutic drugs causes severe side effects, further leading to poor prognosis and patient compliance. Ligand/receptor-mediated targeted drug delivery can improve drug accumulation in the tumor but it always attenuated by protein corona barriers. RESULTS To address these problems, a radically different strategy is proposed that can leave the off-targeted drugs inactive but activate the tumor-distributed drugs for cancer-targeting therapy in a tumor microenvironment-independent manner. The feasibility and effectiveness of this strategy is demonstrated by developing an ultrasound (US)-activated prodrug-loaded liposome (CPBSN38L) comprising the sonosensitizer chlorin e6 (Ce6)-modified lipids and the prodrug of pinacol boronic ester-conjugated SN38 (PBSN38). Once CPBSN38L is accumulated in the tumor and internalized into the cancer cells, under US irradiation, the sonosensitizer Ce6 rapidly induces extensive production of intracellular reactive oxygen species (ROS), thereby initiating a cascade amplified ROS-responsive activation of PBSN38 to release the active SN38 for inducing cell apoptosis. If some of the injected CPBSN38L is distributed into normal tissues, the inactive PBSN38 exerts no pharmacological activity on normal cells. CPBSN38L exhibited strong anticancer activity in multiple murine tumor models of colon adenocarcinoma and hepatocellular carcinoma with no chemotherapy-induced side effects, compared with the standard first-line anticancer drugs irinotecan and topotecan. CONCLUSIONS This study established a side-effect-evitable, universal, and feasible strategy for cancer-targeting therapy.
Collapse
Affiliation(s)
- Yifan Jiang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hongjian Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Tao Lin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiaxin Shen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanan Zhao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wen Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
430
|
Monti M, Ferrari G, Grosso V, Missale F, Bugatti M, Cancila V, Zini S, Segala A, La Via L, Consoli F, Orlandi M, Valerio A, Tripodo C, Rossato M, Vermi W. Impaired activation of plasmacytoid dendritic cells via toll-like receptor 7/9 and STING is mediated by melanoma-derived immunosuppressive cytokines and metabolic drift. Front Immunol 2024; 14:1227648. [PMID: 38239354 PMCID: PMC10795195 DOI: 10.3389/fimmu.2023.1227648] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Plasmacytoid dendritic cells (pDCs) infiltrate a large set of human cancers. Interferon alpha (IFN-α) produced by pDCs induces growth arrest and apoptosis in tumor cells and modulates innate and adaptive immune cells involved in anti-cancer immunity. Moreover, effector molecules exert tumor cell killing. However, the activation state and clinical relevance of pDCs infiltration in cancer is still largely controversial. In Primary Cutaneous Melanoma (PCM), pDCs density decreases over disease progression and collapses in metastatic melanoma (MM). Moreover, the residual circulating pDC compartment is defective in IFN-α production. Methods The activation of tumor-associated pDCs was evaluated by in silico and microscopic analysis. The expression of human myxovirus resistant protein 1 (MxA), as surrogate of IFN-α production, and proximity ligation assay (PLA) to test dsDNA-cGAS activation were performed on human melanoma biopsies. Moreover, IFN-α and CXCL10 production by in vitro stimulated (i.e. with R848, CpG-A, ADU-S100) pDCs exposed to melanoma cell lines supernatants (SN-mel) was tested by intracellular flow cytometry and ELISA. We also performed a bulk RNA-sequencing on SN-mel-exposed pDCs, resting or stimulated with R848. Glycolytic rate assay was performed on SN-mel-exposed pDCs using the Seahorse XFe24 Extracellular Flux Analyzer. Results Based on a set of microscopic, functional and in silico analyses, we demonstrated that the melanoma milieu directly impairs IFN-α and CXCL10 production by pDCs via TLR-7/9 and cGAS-STING signaling pathways. Melanoma-derived immunosuppressive cytokines and a metabolic drift represent relevant mechanisms enforcing pDC-mediated melanoma escape. Discussion These findings propose a new window of intervention for novel immunotherapy approaches to amplify the antitumor innate immune response in cutaneous melanoma (CM).
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgia Ferrari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valentina Grosso
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Nederlands Kanker Instituut, Amsterdam, Netherlands
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Stefania Zini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Agnese Segala
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Consoli
- Oncology Unit, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili di Brescia, Brescia, Italy
| | - Matteo Orlandi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
431
|
Basilicata M, Terrano V, D’Aurelio A, Bruno G, Troiani T, Bollero P, Napolitano S. Oral Adverse Events Associated with BRAF and MEK Inhibitors in Melanoma Treatment: A Narrative Literature Review. Healthcare (Basel) 2024; 12:105. [PMID: 38201012 PMCID: PMC10778825 DOI: 10.3390/healthcare12010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Melanoma cancer represents the most lethal type of skin cancer originating from the malignant transformation of melanocyte cells. Almost 50% of melanomas show the activation of BRAF mutations. The identification and characterization of BRAF mutations led to the development of specific drugs that radically changed the therapeutic approach to melanoma. METHODS We conducted a narrative review of the literature according to a written protocol before conducting the study. This article is based on previously conducted studies. We identified articles by searching electronic databases (Medline, Google Scholar and PubMed). We used a combination of "melanoma", "Braf-Mek inhibitors", " targeted therapy" and "oral side effects". RESULTS Eighteen studies were reported in this article showing the relationship between the use of targeted therapy in melanoma cancer and the development of oral side effects, such as mucositis, hyperkeratosis and cellular proliferation. CONCLUSION Targeted therapy plays an important role in the treatment of melanoma cancer, showing a notable increase in response rate, prolonged progression-free survival and overall survival in BRAF-mutated melanoma patients. Oral side effects represent a common finding over the course of treatment. However, these adverse effects can be easily managed in a multidisciplinary approach involving collaboration between medical oncologists and dental doctors.
Collapse
Affiliation(s)
- Michele Basilicata
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
- UniCamillus-Saint Camillus, International University of Health Sciences, 00131 Rome, Italy
| | - Vincenzo Terrano
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| | - Alessandro D’Aurelio
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
| | - Giovanni Bruno
- Department of Neuroscience, University of Padua, 35121 Padova, Italy
- Department of Industrial Engineering, University of Tor Vergata, 00133 Rome, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| | - Patrizio Bollero
- UOSD Special Care Dentistry, Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, 00133 Rome, Italy; (M.B.); (A.D.); (P.B.)
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (V.T.); (T.T.); (S.N.)
| |
Collapse
|
432
|
Kirkwood JM, Kottschade LA, McWilliams RR, Khushalani NI, Jang S, Hallmeyer S, McDermott DF, Tawbi H, Che M, Lee CH, Ritchings C, Le TK, Park B, Ramsey S. Real-world outcomes with immuno-oncology therapies in advanced melanoma: final results of the OPTIMIzE registry study. Immunotherapy 2024; 16:29-42. [PMID: 37937397 DOI: 10.2217/imt-2022-0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
Aim: The OPTIMIzE registry study evaluated real-world outcomes in patients with advanced melanoma receiving immuno-oncology therapies. Materials and methods: Data were collected for patients treated with anti-programmed death 1 (PD-1) monotherapy (nivolumab or pembrolizumab; n = 147) or nivolumab plus ipilimumab (n = 81) from 2015-2017 and followed for ≥3 years. Results: Nivolumab plus ipilimumab versus anti-PD-1 monotherapy was associated with a nonsignificantly lower risk of death (adjusted HR: 0.83; 95% CI: 0.54-1.28; p = 0.41), higher disease control rate (72 vs 56%; p = 0.04), and stable quality of life, but more grade 3-4 treatment-related adverse events (54 vs 26%; p < 0.0001). Conclusion: These results support the use of immuno-oncology therapy in advanced melanoma.
Collapse
Affiliation(s)
- John M Kirkwood
- Melanoma Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232 USA
| | | | | | - Nikhil I Khushalani
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Sekwon Jang
- Department of Melanoma Research and Therapeutics, Inova Schar Cancer Institute, Fairfax, VA 22031 USA
| | - Sigrun Hallmeyer
- Department of Oncology, Advocate Medical Group, Park Ridge, IL 60068 USA
| | - David F McDermott
- Department of Hematology and Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Min Che
- Bristol Myers Squibb, Princeton, NJ 08540 USA
| | - Cho-Han Lee
- Bristol Myers Squibb, Princeton, NJ 08540 USA
| | | | | | - Boas Park
- Bristol Myers Squibb, Princeton, NJ 08540 USA
| | - Scott Ramsey
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Center, Seattle, WA 98109 USA
| |
Collapse
|
433
|
Du Y, Dai J, Mao L, Wei X, Bai X, Chen L, Lin J, Chi Z, Cui C, Sheng X, Lian B, Tang B, Wang X, Yan X, Li S, Zhou L, Guo J, Chen Y, Si L. Phase Ib study of anlotinib in combination with anti-PD-L1 antibody (TQB2450) in patients with advanced acral melanoma. J Eur Acad Dermatol Venereol 2024; 38:93-101. [PMID: 37625814 DOI: 10.1111/jdv.19467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Acral melanoma, the most common subtype of melanoma in Asians, is often diagnosed at an advanced stage and responds poorly to current programmed cell death protein 1 (PD-1) inhibitors. OBJECTIVES To evaluate the safety and efficacy of TQB2450 and anlotinib in patients with advanced acral melanoma in a phase Ib study (NCT03991975). METHODS Patients received TQB2450 (1200 mg every 3 weeks) and anlotinib (10 mg or 12 mg once daily, 2-week on/1-week off) in the dose-escalation and dose-expansion phases. The primary endpoints were dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and objective response rate (ORR). RESULTS Nineteen patients were enrolled between June 2019 and June 2022. The majority of patients (16 of 19 patients) received anlotinib and TQB2450 as first-line treatment. No DLTs were observed, and MTD was not reached. Eighteen (94.7%) out of 19 patients experienced treatment-related adverse events (TRAEs), but most were grade 1 or 2. Grade 3 or greater TRAEs occurred in seven patients (36.8%). The ORR was 26.3% (two complete responses and three partial responses). The disease control rate was 73.7%. The median duration of response was 30.3 months [95% confidence interval (CI): 5.8-NA]. The median progression-free survival (PFS) was 5.5 months (95% CI: 2.8-NA), and median overall survival was 20.3 months (95% CI: 14.8-NA). Whole-exome sequencing suggested that acquired drug resistance might be attributed to activation of the MAPK signalling pathway and transformation to an immunosuppressive tumour environment. CONCLUSIONS TQB2450 combined with anlotinib showed favourable tolerance and promising anti-tumour activity with a prolonged PFS compared with anti-PD1 monotherapy in patients with advanced acral melanoma.
Collapse
Affiliation(s)
- Yu Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoting Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xue Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ling Chen
- Department of Medical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jing Lin
- Department of Medical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bin Lian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bixia Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xuan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xieqiao Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Siming Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
434
|
Su M, Zhang Z, Jiang P, Wang X, Tong X, Wu G. CAR-T-Cell Therapy Based on Immune Checkpoint Modulation in the Treatment of Hematologic Malignancies. Cell Transplant 2024; 33:9636897241293964. [PMID: 39506457 DOI: 10.1177/09636897241293964] [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: 11/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary immunotherapy that has shown significant success in treating certain hematologic malignancies. However, there are still challenges and limitations associated with this therapy, and not all cancer patients benefit from this therapy alone. Therefore, modifying CAR-T-cell therapy based on immune checkpoints, and its combination with immune checkpoint inhibitors (ICIs), shows promise as a potentially more effective strategy for treating hematologic malignancies. This article outlines the progress of preclinical and clinical trials of CAR-T-cell therapy based on immune checkpoint modulation in the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Manqi Su
- Department of Hematology, Dongyang Hospital Affiliated to WenZhou Medical University, Dongyang People's Hospital, Dongyang, China
| | - Zhanna Zhang
- Department of Hematology, Dongyang Hospital Affiliated to WenZhou Medical University, Dongyang People's Hospital, Dongyang, China
| | - Panruo Jiang
- Department of Hematology, Dongyang Hospital Affiliated to WenZhou Medical University, Dongyang People's Hospital, Dongyang, China
| | - Xiaoxia Wang
- Department of Hematology, Dongyang Hospital Affiliated to WenZhou Medical University, Dongyang People's Hospital, Dongyang, China
| | - Xiangmin Tong
- Affiliated Hangzhou First People's Hospital, School of Medicine, WestLake University, Hangzhou, China
| | - Gongqiang Wu
- Department of Hematology, Dongyang Hospital Affiliated to WenZhou Medical University, Dongyang People's Hospital, Dongyang, China
| |
Collapse
|
435
|
Bravo-Garzón MA, Bornstein-Quevedo L, de Camargo VP, Sanku G, Jansen AM, de Macedo MP, Rico-Restrepo M, Chacón M. BRAF-Mutated Melanoma Journey in Latin America: Expert Recommendations From Diagnosis to Treatment. Cancer Control 2024; 31:10732748241251572. [PMID: 38751033 PMCID: PMC11100406 DOI: 10.1177/10732748241251572] [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: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/19/2024] Open
Abstract
OBJECTIVES • Gather a panel of Latin American experts in testing and treating BRAF-melanoma. • Describe the current landscape of BRAF-mutated melanoma in Latin America. • Outline the current gaps in testing and recommend improvements for testing and treating BRAF-mutated melanoma in the region. INTRODUCTION Melanoma prevalence in Latin America is lower than in high- and middle-income countries. However, recent data indicate that the region's incidence and mortality are rising, with more stage IV patients being diagnosed. According to international clinical practice guidelines, conducting BRAF-mutation testing in patients with stage III or stage IV melanoma and high-risk resected disease is imperative. Still, BRAF-mutation testing and targeted therapies are inconsistently available in the region. METHODS Americas Health Foundation convened a meeting of Latin American experts on BRAF-mutated melanoma to develop guidelines and recommendations for diagnosis through treatment. RESULTS AND CONCLUSIONS Some recommendations for improving diagnostics through improving access and reducing the cost of BRAF-mutation testing, enhancing efficiency in pathology laboratories, and creating country-specific local guidelines. The panel also gave treatment recommendations for neo-adjuvant therapy, adjuvant therapy, and therapy for patients with metastatic disease in Latin America.
Collapse
Affiliation(s)
- María A. Bravo-Garzón
- Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center – CTIC, Bogotá D.C., Colombia
| | | | | | | | | | | | | | - Matías Chacón
- Alexander Fleming Institute, Buenos Aires, Argentina
| |
Collapse
|
436
|
Hepner A, Versluis JM, Wallace R, Allayous C, Brown LJ, Trojaniello C, Gerard CL, Jansen YJ, Bhave P, Neyns B, Haydon A, Michielin O, Mangana J, Klein O, Shoushtari AN, Warner AB, Ascierto PA, McQuade JL, Carlino MS, Zimmer L, Lebbe C, Johnson DB, Sandhu S, Atkinson V, Blank CU, Lo SN, Long GV, Menzies AM. The features and management of acquired resistance to PD1-based therapy in metastatic melanoma. Eur J Cancer 2024; 196:113441. [PMID: 37988842 PMCID: PMC11270881 DOI: 10.1016/j.ejca.2023.113441] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Anti-PD-1 therapy (PD1) either alone or with anti-CTLA-4 (CTLA4), has high initial response rates, however 20% of patients (pts) with complete response (CR) and 30% with partial response (PR) within 12 months of treatment experience subsequent disease progression by 6 years. The nature and optimal management of this acquired resistance (AR) remains unknown. METHODS Pts from 16 centres who responded to PD1-based therapy and who later progressed were examined. Demographics, disease characteristics and subsequent treatments were evaluated. RESULTS 299 melanoma pts were identified, median age 64y, 44% BRAFV600m. 172 (58%) received PD1 alone, 114 (38%) PD1/CTLA4 and 13 (4%) PD1 and an investigational drug. 90 (30%) pts had CR, 209 (70%) PR. Median time to AR was 12.6 mo (95% CI, 11.3, 14.2). Most (N = 193, 65%) progressed in a single organ site, and in a solitary lesion (N = 151, 51%). The most frequent sites were lymph nodes (38%) and brain (25%). Management at AR included systemic therapy (ST, 45%), local therapy (LT) +ST (31%), LT alone (21%), or observation (3%). There was no statistical difference in PFS2 or OS based on management, however, PFS2 was numerically superior for pts treated with ST alone who progressed off PD1 therapy than those who progressed on PD1 (2-year PFS2 42% versus 25%, p = 0.249). mOS from AR was 38.0 months (95% CI, 29.5-NR); longer in single-site versus multi-site progression (2-year OS 70% vs 54%, p < 0·001). CONCLUSIONS Acquired resistance to PD1 therapy in melanoma is largely oligometastatic, and pts may have a favorable survival outcome following salvage treatment.
Collapse
Affiliation(s)
- Adriana Hepner
- Melanoma Institute Australia, The University of Sydney, NSW, Australia; Instituto do Cancer do Estado de Sao Paulo, SP, Brazil
| | | | - Roslyn Wallace
- Sir Peter MacCallum Cancer Centre Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Clara Allayous
- Université Paris Cite, Dermato-Oncology AP-HP Hôpital Saint Louis, INSERM U976, F-75010 Paris, France
| | - Lauren Julia Brown
- Crown Princess Mary Cancer Centre Westmead and Blacktown Hospitals, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | | | - Camille Lea Gerard
- Precision Oncology Center Oncology department, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Yanina Jl Jansen
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven 3000, Belgium
| | - Prachi Bhave
- Sir Peter MacCallum Cancer Centre Department of Oncology, The University of Melbourne, Melbourne, Australia; Department of Medical Oncology, Alfred Health, Melbourne, Australia
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Andrew Haydon
- Department of Medical Oncology, Alfred Health, Melbourne, Australia; Monash University, Melbourne, Australia
| | - Olivier Michielin
- Precision Oncology Center Oncology department, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | | | - Oliver Klein
- Olivia Newton-John Cancer Centre and Austin Health, Melbourne, Australia
| | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Celeste Lebbe
- Université Paris Cite, Dermato-Oncology AP-HP Hôpital Saint Louis, INSERM U976, F-75010 Paris, France
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville TN, USA
| | - Shahneen Sandhu
- Sir Peter MacCallum Cancer Centre Department of Oncology, The University of Melbourne, Melbourne, Australia; University of Melbourne, Melbourne, Australia
| | - Victoria Atkinson
- University of Queensland and Princess Alexandra and Greenslopes Private Hospital, Brisbane, Australia
| | - Christian U Blank
- Netherlands Cancer Institute (NKI), Amsterdam, the Netherlands; Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, NSW, Australia.
| |
Collapse
|
437
|
Guven DC, Aykan MB, Muglu H, Bayram E, Helvaci K, Dursun B, Celayir M, Chelebiyev E, Nayir E, Erman M, Sezer A, Urun Y, Demirci U, Er O, Disel U, Bilici A, Arslan C, Karadurmus N, Kilickap S. The efficacy of immunotherapy and chemoimmunotherapy in patients with advanced rare tumors: A Turkish oncology group (TOG) study. Cancer Med 2024; 13:e6869. [PMID: 38140782 PMCID: PMC10809296 DOI: 10.1002/cam4.6869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
INTRODUCTION The advances in immune checkpoint inhibitors (ICIs) were relatively slow in rare tumors. Therefore, we conducted a multi-center study evaluating the efficacy of ICI monotherapy and the combination of ICIs with chemotherapy (CT) in patients with advanced rare tumors. METHODS In this retrospective cohort study, we included 93 patients treated with ICIs for NCI-defined rare tumors from the 12 cancer centers in Turkey. The primary endpoints were the overall response (ORR) and disease control rate (DCR). RESULTS The cohort's median age was 56, and 53.8% of the patients were male. The most frequent diagnosis was sarcoma (29%), and 81.7% of the patients were previously treated with at least one line of systemic therapy in the advanced stage. The ORR and DCR were 36.8% and 63.2%, respectively. The germ cell tumors had the lowest ORR (0%), while the Merkel cell carcinoma had the highest ORR to ICIs (57.1%). Patients treated with ICI + ICI or ICI plus chemotherapy combinations had higher ORR (55.2% vs. 27.6%, p = 0.012) and DCR (82.8% vs. 53.4%, p = 0.008). The median OS was 13.47 (95% CI: 7.79-19.15) months, and the six and 12-month survival rates were 71% and 52%. The median duration of response was 16.59 months, and the 12-month progression-free survival rate was 66% in responders. The median time-to-treatment failure was 5.06 months (95% CI: 3.42-6.71). Three patients had high-grade irAEs with ICIs (grade 3 colitis, grade 3 gastritis, and grade 3 encephalitis in one patient each). CONCLUSION We observed over 30% ORR and a 13-month median OS in patients with rare cancers treated with ICI monotherapy or ICI plus CT combinations. The response rates to ICIs or ICIs plus CT significantly varied across different tumor types. Responding patients had over 2 years of survival, highlighting a need for further trials with ICIs for patients with rare tumors.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Musa Baris Aykan
- Department of Medical OncologyGulhane School of Medicine, University of Health SciencesAnkaraTurkey
| | - Harun Muglu
- Istanbul Medipol University Faculty of MedicineIstanbulTurkey
| | - Ertugrul Bayram
- Department of Medical OncologyCukurova UniversityAdanaTurkey
| | | | - Bengü Dursun
- Department of Medical OncologyAnkara UniversityAnkaraTurkey
| | - Melisa Celayir
- Department of Medical OncologyMAA Acıbadem UniversityİstanbulTurkey
| | - Elvin Chelebiyev
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Erdinc Nayir
- Department of Medical OncologyMersin Medical Park HospitalMersinTurkey
| | - Mustafa Erman
- Department of Medical OncologyHacettepe University Cancer InstituteAnkaraTurkey
| | - Ahmet Sezer
- Baskent University Adana HospitalAdanaTurkey
| | - Yuksel Urun
- Department of Medical OncologyAnkara UniversityAnkaraTurkey
| | | | - Ozlem Er
- Department of Medical OncologyMAA Acıbadem UniversityİstanbulTurkey
| | - Umut Disel
- Department of Medical OncologyAcibadem Adana HospitalAdanaTurkey
| | - Ahmet Bilici
- Istanbul Medipol University Faculty of MedicineIstanbulTurkey
| | - Cagatay Arslan
- Department of Medical OncologySchool of Medicine, Medical Park Hospital, Izmir Economy UniversityIzmirTurkey
| | - Nuri Karadurmus
- Department of Medical OncologyGulhane School of Medicine, University of Health SciencesAnkaraTurkey
| | - Saadettin Kilickap
- Department of Medical OncologyIstinye University Faculty of MedicineIstanbulTurkey
| |
Collapse
|
438
|
Zhao C, Yu M, Li Y. Pan-cancer analysis reveals the pro-oncogenic role of N6-methyladenosine (m6A)-regulated NTMT1 in head and neck squamous cell carcinoma. J Biochem Mol Toxicol 2024; 38:e23603. [PMID: 38014887 DOI: 10.1002/jbt.23603] [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: 02/14/2023] [Revised: 09/09/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Head and neck squamous cell carcinoma (HNSC) is a common and fatal tumor with a bleak prognosis, posing a significant threat to human health. N6-methyladenosine (m6A) modification regulates tumor progression by modulating gene expression post-transcriptionally. Nevertheless, the specific function of m6A-modified tumor drivers in HNSC remains largely uncharted. In this study, we revealed the pro-oncogenic role of m6A-regulated NTMT1 in HNSC through comprehensive pan-cancer analysis and experimental validation. By scrutinizing the prognostic and expression profiles of NTMT1 across over 30 cancer types, we observed a significant association between NTMT1 and patient overall survival in ACC, HNSC, LAML, LGG, KIRC, and STAD. Moreover, we find a close correlation between NTMT1 and disease-free survival in ACC, HNSC, LUSC, UVM, KIRC, and STAD. NTMT1 exhibited dysregulation in 15 cancers, including CESC, CHOL, COAD, DLBC, GBM, HNSC, LGG, LIHC, PAAD, READ, SKCM, THYM, UCS, LAML, and TGCT. Integrated data underscored the critical involvement of NTMT1 in HNSC. Furthermore, the expression of NTMT1 was closely associated with tumor stage and immune infiltration in HNSC. Functionally, NTMT1 deficiency was demonstrated to significantly impede cell proliferation and cell-cycle progression in HNSC. Mechanistically, METTL3 was elucidated to mediate the epigenetic upregulation of NTMT1 in HNSC in an m6A-dependent manner, and the overexpression of METTL3 was shown to alleviate the inhibitory impact of downregulated NTMT1 on HNSC proliferation. In conclusion, our findings enhance our understanding of NTMT1's role across various cancer types and offer a rationale for clinically targeting NTMT1 as a therapeutic approach for HNSC.
Collapse
Affiliation(s)
- Chunhong Zhao
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Min Yu
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yujie Li
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
439
|
Ceci C, García-Chico C, Atzori MG, Lacal PM, Lista S, Santos-Lozano A, Graziani G, Pinto-Fraga J. Impact of Physical Exercise on Melanoma Hallmarks: Current Status of Preclinical and Clinical Research. J Cancer 2024; 15:1-19. [PMID: 38164270 PMCID: PMC10751671 DOI: 10.7150/jca.88559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 01/03/2024] Open
Abstract
In recent years, accumulating evidence from preclinical and clinical studies consistently indicated that physical activity/exercise plays a crucial role in reducing the incidence and recurrence of various malignancies, by exerting a beneficial modulation of cancer hallmarks. Moreover, physical activity is suggested to attenuate certain adverse effects of anticancer therapy, including the reduction of cardiovascular toxicity and symptoms related to depression and anxiety, among others, while preserving muscular strength. In the case of melanoma, the relationship with physical activity has been critically debated. Historically, several cohort studies and meta-analyses reported a positive association between physical activity/exercise and melanoma risk. This association was primarily attributed to outdoor activities that may expose the skin to UV radiation, a well-known risk factor for melanocyte transformation. However, more recent evidence does not support such association and recognizes physical activity/exercise role in both melanoma prevention and progression. Nevertheless, sun protection is recommended during outdoor training to minimize UV radiation exposure. This narrative review summarizes preclinical and clinical data about physical activity effects on melanoma hallmarks. Specifically, experimental evidence is reported concerning (i) invasion and metastasis, (ii) reprogramming of energy metabolism, (iii) angiogenesis, (iv) resistance to cell death, (v) evasion from immune destruction, and (vi) tumor-promoting inflammation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Celia García-Chico
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | | | | | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - José Pinto-Fraga
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| |
Collapse
|
440
|
Fedele D, Moroso S, Turoldo A, Bazzocchi G, Conforti C, Zalaudek I, Guglielmi A. A Dramatic Response to Second-Line Nivolumab and Ipilimumab in BRAF-V600-Mutated Metastatic Melanoma. Case Rep Oncol 2024; 17:161-168. [PMID: 38288458 PMCID: PMC10824524 DOI: 10.1159/000535902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/13/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Current treatment options for BRAF V600-mutated unresectable stage III/IV melanoma include anti-PD-1 monotherapy or combination with anti-CTLA-4 or anti-LAG-3 agents, BRAF/MEK inhibitors, and clinical trials. The strategy of combination immunotherapy with nivolumab and ipilimumab has shown promising results, achieving higher response rates, longer duration of response, improved progression-free survival, and enhanced overall survival. The optimal sequence of treatments remains a topic of interest, with preliminary data suggesting a greater effectiveness of immunotherapy as the first-line approach. Preclinical trials have indicated that the efficacy of this sequence may be due to the modification of the immune environment by BRAF kinase inhibitors, leading to immune escape by tumor cells and resistance to immune checkpoint inhibitors. Case Presentation We present a case of a 72-year-old woman with high-burden metastatic melanoma who failed to respond to prior targeted therapy with BRAF/MEK inhibitors and exhibited a successful response to the second-line treatment with ipilimumab and nivolumab. We discuss the potential reasons for this positive outcome contributing to the current debate concerning treatment sequences, resistance mechanisms, and biomarkers predictive of response to immune checkpoint inhibitors in metastatic melanoma. Conclusion We believe that in few years the therapeutic algorithms in BRAF V600-mutated unresectable stage III/IV melanoma will be more complex since they will define clearly the correct therapeutic sequences with the inclusion of new immune checkpoint inhibitor drugs and multiple predictive biomarkers of response to better select patients eligible to immunotherapy.
Collapse
Affiliation(s)
- Dahlia Fedele
- Department of Medical Oncology, ASUGI, Maggiore Hospital, Trieste, Italy
| | - Stefano Moroso
- Department of Medical Oncology, ASUGI, Maggiore Hospital, Trieste, Italy
| | - Angelo Turoldo
- University Department of Clinical Surgery, ASUGI, Cattinara Hospital, Trieste, Italy
| | - Gabriele Bazzocchi
- Department of Diagnostic Imaging, ASUGI, Maggiore Hospital, Trieste, Italy
| | | | - Iris Zalaudek
- University Department of Clinical Dermatology, ASUGI, Maggiore Hospital, Trieste, Italy
| | | |
Collapse
|
441
|
Malekzadeh P, Brady MS. Adjuvant therapy for stage IIB + IIC melanoma. J Surg Oncol 2024; 129:91-96. [PMID: 38031847 DOI: 10.1002/jso.27534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Affiliation(s)
- Parisa Malekzadeh
- Department of Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mary S Brady
- Department of Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
442
|
Várvölgyi T, Janka EA, Szász I, Koroknai V, Toka-Farkas T, Szabó IL, Ványai B, Szegedi A, Emri G, Balázs M. Combining Biomarkers for the Diagnosis of Metastatic Melanoma. J Clin Med 2023; 13:174. [PMID: 38202181 PMCID: PMC10779676 DOI: 10.3390/jcm13010174] [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: 11/09/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The early detection of melanoma relapse can improve patient survival; thus, there is a great need for easily accessible biomarkers that facilitate the diagnosis of metastatic disease. We investigated the diagnostic effect of blood biomarkers such as lactate dehydrogenase (LDH), S100B, and osteopontin in the detection of metastases. Clinical data and peripheral blood samples of 206 melanoma patients were collected (no metastasis, N = 120; metastasis, N = 86). The discriminative power of blood biomarkers, patient demographics, and clinicopathological parameters of primary melanomas were evaluated using univariate and multivariate logistic regression models and receiver operating characteristic (ROC) analysis to determine the area under the curve (AUC). Plasma osteopontin levels showed a significant and independent effect on the probability of metastasis, similar to serum S100B levels. In addition, the location of the primary tumor on the lower extremities and the American Joint Committee on Cancer (AJCC) categories pT2b-3a, pT3b-4a, and pT4b were associated with the diagnosis of metastasis. Importantly, the combination of the three blood biomarkers and primary tumor localization and AJCC pT category yielded excellent discrimination (AUC: training set: 0.803; validation set: 0.822). In conclusion, plasma osteopontin can be classified as a melanoma biomarker; moreover, by combining clinicopathological prognostic variables, the diagnostic effect of blood biomarkers in the detection of metastatic melanoma can be improved.
Collapse
Affiliation(s)
- Tünde Várvölgyi
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
| | - Eszter Anna Janka
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - István Szász
- HUN-REN-UD Public Health Research Group, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (V.K.)
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary
| | - Viktória Koroknai
- HUN-REN-UD Public Health Research Group, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (V.K.)
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary
| | - Tünde Toka-Farkas
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
| | - Imre Lőrinc Szabó
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Beatrix Ványai
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
| | - Andrea Szegedi
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Gabriella Emri
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (E.A.J.); (T.T.-F.); (I.L.S.); (B.V.); (A.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Margit Balázs
- HUN-REN-UD Public Health Research Group, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (V.K.)
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary
| |
Collapse
|
443
|
Noujarède J, Carrié L, Garcia V, Grimont M, Eberhardt A, Mucher E, Genais M, Schreuder A, Carpentier S, Ségui B, Nieto L, Levade T, Puig S, Torres T, Malvehy J, Harou O, Lopez J, Dalle S, Caramel J, Gibot L, Riond J, Andrieu-Abadie N. Sphingolipid paracrine signaling impairs keratinocyte adhesion to promote melanoma invasion. Cell Rep 2023; 42:113586. [PMID: 38113139 DOI: 10.1016/j.celrep.2023.113586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/20/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer due to its propensity to metastasize. It arises from melanocytes, which are attached to keratinocytes within the basal epidermis. Here, we hypothesize that, in addition to melanocyte-intrinsic modifications, dysregulation of keratinocyte functions could initiate early-stage melanoma cell invasion. We identified the lysolipid sphingosine 1-phosphate (S1P) as a tumor paracrine signal from melanoma cells that modifies the keratinocyte transcriptome and reduces their adhesive properties, leading to tumor invasion. Mechanistically, tumor cell-derived S1P reduced E-cadherin expression in keratinocytes via S1P receptor dependent Snail and Slug activation. All of these effects were blocked by S1P2/3 antagonists. Importantly, we showed that epidermal E-cadherin expression was inversely correlated with the expression of the S1P-producing enzyme in neighboring tumors and the Breslow thickness in patients with early-stage melanoma. These findings support the notion that E-cadherin loss in the epidermis initiates the metastatic cascade in melanoma.
Collapse
Affiliation(s)
- Justine Noujarède
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Lorry Carrié
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Virginie Garcia
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Maxime Grimont
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France
| | - Anaïs Eberhardt
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France; Service de Dermatologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Elodie Mucher
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Matthieu Genais
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Anne Schreuder
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Stéphane Carpentier
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Bruno Ségui
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Laurence Nieto
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Thierry Levade
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France; Laboratoire de Biochimie Métabolique, CHU de Toulouse, Toulouse, France
| | - Susana Puig
- Melanoma Unit, Department of Dermatology, University of Barcelona, Barcelona, Spain & CIBER of Rare Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Teresa Torres
- Melanoma Unit, Department of Dermatology, University of Barcelona, Barcelona, Spain & CIBER of Rare Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Josep Malvehy
- Melanoma Unit, Department of Dermatology, University of Barcelona, Barcelona, Spain & CIBER of Rare Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Olivier Harou
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France; Service de Dermatologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Jonathan Lopez
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France; Service de Dermatologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Stéphane Dalle
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France; Service de Dermatologie, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Pierre Bénite, France
| | - Julie Caramel
- Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre de Recherches en Cancérologie de Lyon, Lyon, France
| | - Laure Gibot
- Université Toulouse III Paul-Sabatier, Laboratoire des Interactions Moléculaires et Réactivité Chimique et Photochimique, CNRS UMR5623, Toulouse, France
| | - Joëlle Riond
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Nathalie Andrieu-Abadie
- Université de Toulouse, INSERM, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France.
| |
Collapse
|
444
|
Poletto S, Paruzzo L, Nepote A, Caravelli D, Sangiolo D, Carnevale-Schianca F. Predictive Factors in Metastatic Melanoma Treated with Immune Checkpoint Inhibitors: From Clinical Practice to Future Perspective. Cancers (Basel) 2023; 16:101. [PMID: 38201531 PMCID: PMC10778365 DOI: 10.3390/cancers16010101] [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: 10/10/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The introduction of immunotherapy revolutionized the treatment landscape in metastatic melanoma. Despite the impressive results associated with immune checkpoint inhibitors (ICIs), only a portion of patients obtain a response to this treatment. In this scenario, the research of predictive factors is fundamental to identify patients who may have a response and to exclude patients with a low possibility to respond. These factors can be host-associated, immune system activation-related, and tumor-related. Patient-related factors can vary from data obtained by medical history (performance status, age, sex, body mass index, concomitant medications, and comorbidities) to analysis of the gut microbiome from fecal samples. Tumor-related factors can reflect tumor burden (metastatic sites, lactate dehydrogenase, C-reactive protein, and circulating tumor DNA) or can derive from the analysis of tumor samples (driver mutations, tumor-infiltrating lymphocytes, and myeloid cells). Biomarkers evaluating the immune system activation, such as IFN-gamma gene expression profile and analysis of circulating immune cell subsets, have emerged in recent years as significantly correlated with response to ICIs. In this manuscript, we critically reviewed the most updated literature data on the landscape of predictive factors in metastatic melanoma treated with ICIs. We focus on the principal limits and potentiality of different methods, shedding light on the more promising biomarkers.
Collapse
Affiliation(s)
- Stefano Poletto
- Department of Oncology, University of Turin, AOU S. Luigi Gonzaga, 10043 Orbassano, Italy;
| | - Luca Paruzzo
- Department of Oncology, University of Turin, 10124 Turin, Italy; (L.P.); (D.S.)
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessandro Nepote
- Department of Oncology, University of Turin, AOU S. Luigi Gonzaga, 10043 Orbassano, Italy;
| | - Daniela Caravelli
- Medical Oncology Division, Candiolo Cancer Institute, FPO-IRCCs, 10060 Candiolo, Italy; (D.C.); (F.C.-S.)
| | - Dario Sangiolo
- Department of Oncology, University of Turin, 10124 Turin, Italy; (L.P.); (D.S.)
| | | |
Collapse
|
445
|
Spiliopoulou P, Holanda Lopes CD, Spreafico A. Promising and Minimally Invasive Biomarkers: Targeting Melanoma. Cells 2023; 13:19. [PMID: 38201222 PMCID: PMC10777980 DOI: 10.3390/cells13010019] [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: 11/06/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
The therapeutic landscape of malignant melanoma has been radically reformed in recent years, with novel treatments emerging in both the field of cancer immunotherapy and signalling pathway inhibition. Large-scale tumour genomic characterization has accurately classified malignant melanoma into four different genomic subtypes so far. Despite this, only somatic mutations in BRAF oncogene, as assessed in tumour biopsies, has so far become a validated predictive biomarker of treatment with small molecule inhibitors. The biology of tumour evolution and heterogeneity has uncovered the current limitations associated with decoding genomic drivers based only on a single-site tumour biopsy. There is an urgent need to develop minimally invasive biomarkers that accurately reflect the real-time evolution of melanoma and that allow for streamlined collection, analysis, and interpretation. These will enable us to face challenges with tumour tissue attainment and process and will fulfil the vision of utilizing "liquid biopsy" to guide clinical decisions, in a manner akin to how it is used in the management of haematological malignancies. In this review, we will summarize the most recent published evidence on the role of minimally invasive biomarkers in melanoma, commenting on their future potential to lead to practice-changing discoveries.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | | | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
| |
Collapse
|
446
|
Murphy ML, Rogers D. Tuberculosis reactivation demonstrated by choroiditis and inflammatory choroidal neovascular membrane in a patient treated with immune checkpoint inhibitors for malignant mucosal melanoma. J Ophthalmic Inflamm Infect 2023; 13:54. [PMID: 38110833 PMCID: PMC10728377 DOI: 10.1186/s12348-023-00374-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/19/2023] [Indexed: 12/20/2023] Open
Abstract
PURPOSE To describe a complex case of ocular tuberculosis reactivation with anterior uveitis, choroiditis and inflammatory choroidal neovascular membrane (CNVM) following immune checkpoint inhibitor (ICPI) treatment of malignant mucosal melanoma. METHODS A retrospective collection of medical history, clinical findings and multimodal imaging with literature review of the topic was conducted. RESULTS A 52-year-old Romanian female developed reduced vision and photophobia after three cycles of ICPI therapy comprised of ipilimumab and nivolumab. Bilateral anterior uveitis, multiple left eye choroidal lesions and a CNVM were confirmed using slit-lamp examination with ancillary multimodal imaging. Retinal changes in the right eye as well as a history of previously treated posterior uveitis and high-risk ethnicity increased clinical suspicion for ocular tuberculosis (TB) reactivation. The diagnosis was confirmed by TB positivity on polymerase chain reaction (PCR) analysis of lung aspirate followed by significant clinical improvement on systemic anti-tubercular therapy (ATT), systemic steroids and anti-vascular endothelial growth factor (VEGF) therapy. CONCLUSIONS ICPIs can cause a myriad of ocular issues, both by primary immunomodulatory effects as well as secondary reactivation of latent disease.
Collapse
Affiliation(s)
- Melissa L Murphy
- School of Medicine and Medical Sciences, Mater Misericordiae University Hospital, Eccles Street Dublin, 7, Dublin, Ireland.
| | - Duncan Rogers
- School of Medicine and Medical Sciences, Mater Misericordiae University Hospital, Eccles Street Dublin, 7, Dublin, Ireland
| |
Collapse
|
447
|
Abstract
Abstract
Mucosal melanoma (MM) is extremely rare in Caucasians, whereas it is the second predominant melanoma subtype in Asian and other non-Caucasian populations. Distinct from cutaneous melanoma in terms of epidemiology, biology, and molecular characteristics, MM is characterized by more aggressive biological behavior, lower mutational burden, more chromosomal structure variants, and poorer prognosis. Because of the rarity of MM, its biological features are not fully understood, and potential novel therapies are less well depicted. Whereas immunotherapy has shown encouraging efficacy for cutaneous melanoma, its efficacy in MM is unclear due to limited sample sizes in clinical trials. Thus, in this review, we describe the epidemiological, clinical, and molecular features of MM and summarize the efficacies of different immunotherapies for MM, including immune checkpoint inhibitors, vaccines, oncolytic virus therapy, adoptive T-cell therapy, and various combination therapies.
Collapse
|
448
|
Pepys J, Stoff R, Ramon-Gonen R, Ben-Betzalel G, Grynberg S, Frommer RS, Schachter J, Asher N, Taliansky A, Nikitin V, Dori A, Shelly S. Incidence and Outcome of Neurologic Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors in Patients With Melanoma. Neurology 2023; 101:e2472-e2482. [PMID: 37652699 PMCID: PMC10791056 DOI: 10.1212/wnl.0000000000207632] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/30/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Neurologic immune-related adverse events (n-irAEs) reportedly occur in up to 8% of patients treated with immune checkpoint inhibitors (ICIs) of all age groups. We investigated the association between age and n-irAEs in patients treated with ICIs and examined the effect of n-irAEs on survival outcomes in a large cohort of patients with melanoma. METHODS We conducted a retrospective analysis of patients with advanced melanoma treated with ICIs at Ella Institute for Immuno-oncology and Melanoma between January 1, 2015, and April 20, 2022. The outcomes of interest were defined as the investigation of age-related frequency and severity of n-irAEs, the need for ICI interruption, the treatment required for n-irAE management, the safety of ICI reintroduction, and n-irAE's effect on survival. RESULTS ICI was administered to 937 patients. At least one irAE occurred in 73.5% (n = 689) of them. Among the study population, 8% (n = 76) developed a n-irAE, with a median age of 66 years in female and 68 years in male patients at onset. The median follow-up after n-irAE was 1,147 days (IQR: 1,091.5 range: 3,938). Fewer irAEs occurred in patients older than 70 years (median: 3 events, p = 0.04, CI 2.5-4.7) while specifically colitis and pneumonitis were more common in the 18-60 age group (p = 0.03, 95% CI 0.8-0.38, p = 0.009, 95% CI 0.06-0.2). Grade ≥ 3 toxicity was seen in 35.5% of patients across age groups. The median time from ICI administration to n-irAE development was 48 days across age groups. Common n-irAE phenotypes were myositis (44.7%), encephalitis (10.5%), and neuropathy (10.5%). N-irAE required hospitalization in 40% of patients and steroids treatment in 46% with a median of 4 days from n-irAE diagnosis to steroids treatment initiation. Nine patients needed second-line immunosuppressive treatment. Rechallenge did not cause additional n-irAE in 71% of patients. Developing n-irAE (HR = 0.4, 95% CI 0.32-0.77) or any irAE (HR = 0.7195% CI 0.56-0.88) was associated with longer survival. DISCUSSION N-irAEs are a relatively common complication of ICIs (8% of our cohort). Older age was not associated with its development or severity, in contrast with non-n-irAEs which occurred less frequently in the elderly population. Rechallenge did not result in life-threatening AEs. Development of any irAEs was associated with longer survival; this association was stronger with n-irAEs.
Collapse
Affiliation(s)
- Jack Pepys
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronen Stoff
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Roni Ramon-Gonen
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Guy Ben-Betzalel
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Shirly Grynberg
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronnie Shapira Frommer
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Jacob Schachter
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Nethanel Asher
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Alisa Taliansky
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Vera Nikitin
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Amir Dori
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| | - Shahar Shelly
- From the Department of Biomedical Sciences (J.P.), Humanitas University, Milan, Italy; Ella Institute for Immuno-Oncology and Melanoma (R.S., G.B.-B., S.G., R.S.F., J.S., N.A.), Sheba Medical Center; Department of Neurology and Neurosurgery (R.S., G.B.-B., S.G., R.S.F., J.S., N.A., A.T., V.N., A.D.), Sackler Faculty of Medicine, Tel Aviv University; The Graduate School of Business Administration (R.R.-G.), Bar-Ilan University; Department of Neurology (A.T., V.N., A.D.), Sheba Medical Center, Ramat-Gan, Israel; Department of Neurology Mayo Clinic (S.S.), Rochester, MN; Department of Neurology (S.S.), Rambam Medical Center; and Rappaport Faculty of Medicine (S.S.), Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
449
|
Galligan A, Wallace R, Krishnamurthy B, Kay TWH, Sachithanandan N, Chiang C, Sandhu S, Hicks RJ, Iravani A. Increased Thyroidal Activity on Routine FDG-PET/CT after Combination Immune Checkpoint Inhibition: Temporal Associations with Clinical and Biochemical Thyroiditis. Cancers (Basel) 2023; 15:5803. [PMID: 38136348 PMCID: PMC10741830 DOI: 10.3390/cancers15245803] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND FDG-PET/CT used for immune checkpoint inhibitor (ICI) response assessment can incidentally identify immune-related adverse events (irAEs), including thyroiditis. This study aimed to correlate the time course of FDG-PET/CT evidence of thyroiditis with clinical and biochemical evolution of thyroid dysfunction. METHODS A retrospective review was performed by two independent blinded nuclear medicine physicians (NMPs) of thyroidal FDG uptake in 127 patients who underwent PET/CT between January 2016 and January 2019 at baseline and during treatment monitoring of combination ICI therapy for advanced melanoma. Interobserver agreement was assessed and FDG-PET/CT performance defined by a receiver-operating characteristic (ROC) curve using thyroid function tests (TFTs) as the standard of truth. Thyroid maximum standardized uptake value (SUVmax) and its temporal changes with respect to the longitudinal biochemistry were serially recorded. RESULTS At a median of 3 weeks after commencing ICI, 43/127 (34%) had a diagnosis of thyroiditis established by abnormal TFTs. FDG-PET/CT was performed at baseline and at a median of 11 weeks (range 3-32) following the start of therapy. ROC analysis showed an area under the curve of 0.87 (95% CI 0.80, 0.94) for FDG-PET/CT for detection of thyroiditis with a positive predictive value of 93%. Among patients with biochemical evidence of thyroiditis, those with a positive FDG-PET/CT were more likely to develop overt hypothyroidism (77% versus 35%, p < 0.01). In the evaluation of the index test, there was an almost perfect interobserver agreement between NMPs of 93.7% (95% CI 89.4-98.0), kappa 0.83. CONCLUSION Increased metabolic activity of the thyroid on routine FDG-PET/CT performed for tumoral response of patients undergoing ICI therapy is generally detected well after routine biochemical diagnosis. Elevation of FDG uptake in the thyroid is predictive of overt clinical hypothyroidism and suggests that an ongoing robust inflammatory response beyond the initial thyrotoxic phase may be indicative of thyroid destruction.
Collapse
Affiliation(s)
- Anna Galligan
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Roslyn Wallace
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (R.W.); (S.S.)
| | - Balasubramanian Krishnamurthy
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Thomas W. H. Kay
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Nirupa Sachithanandan
- Department of Endocrinology and Diabetes, St Vincent’s Hospital Melbourne, Melbourne, VIC 3065, Australia; (B.K.); (T.W.H.K.); (N.S.)
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
- Department of Internal Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
| | - Cherie Chiang
- Department of Internal Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Medicine, Royal Melbourne Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (R.W.); (S.S.)
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Rodney J. Hicks
- Department of Medicine, St Vincent’s Hospital Medical School, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Amir Iravani
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
450
|
Minor DR, Kim KB, Karuturi RKM, Kashani-Sabet M. Extended long-term follow-up of metastatic melanoma patients treated with immunotherapy: late relapses and second primary melanomas. Front Oncol 2023; 13:1241917. [PMID: 38111529 PMCID: PMC10725969 DOI: 10.3389/fonc.2023.1241917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023] Open
Abstract
Background Immunotherapy has revolutionized the treatment of patients with advanced melanoma as well as other cancers. Most studies, whether of interleukin-2 or checkpoint inhibitor therapies, have limited follow-up after 5 years, making the incidence of late relapses uncertain. In addition, the incidence of second primary melanomas in patients with stage IV melanoma treated with immunotherapy has rarely been reported. Methods We performed a single-institution retrospective study of stage IV melanoma patients treated with interleukin-2 or checkpoint inhibitors over the period from 1992 to 2013. We found 59 patients alive and in remission 5 years after the beginning of immunotherapy and reviewed their subsequent clinical course. Results This 59-patient cohort had a median follow-up of 13.1 years, with 36 patients followed up for at least 10 years. Four patients (6.8%) had relapses of their metastatic melanoma at 5, 8, 15, and 17 years after starting the successful immunotherapy. Three of the four are still alive. Only one patient in 690 patient-years of observation had a second primary invasive melanoma. Conclusion Although late relapses after immunotherapy for melanoma do occur, we can conclude that the prognosis of stage IV melanoma patients in continuous remission 5 years after starting immunotherapy is excellent, with a progression-free survival of approximately 85% and a melanoma-specific survival of approximately 95% at 20 years in our series. Our incidence of second primary melanomas is lower than usually reported. These results have important implications regarding the follow-up of stage IV melanoma patients successfully treated with immunotherapy.
Collapse
Affiliation(s)
- David R. Minor
- California Pacific Medical Center Research Institute and Center for Melanoma Research and Treatment, San Francisco, CA, United States
| | - Kevin B. Kim
- California Pacific Medical Center Research Institute and Center for Melanoma Research and Treatment, San Francisco, CA, United States
| | | | - Mohammed Kashani-Sabet
- California Pacific Medical Center Research Institute and Center for Melanoma Research and Treatment, San Francisco, CA, United States
| |
Collapse
|