1
|
Chen J, Tarantino G, Severgnini M, Baginska J, Giobbie-Hurder A, Weirather JL, Manos M, Russell JD, Pfaff KL, Rodig SJ, Huang AY, Brennick R, Nazzaro M, Hathaway E, Holovatska M, Manuszak C, Ranasinghe S, Liu D, Hodi FS. Circulating cytokine associations with clinical outcomes in melanoma patients treated with combination nivolumab plus ipilimumab. Oncoimmunology 2025; 14:2432723. [PMID: 39699928 DOI: 10.1080/2162402x.2024.2432723] [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: 07/30/2024] [Revised: 10/18/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Nivolumab plus ipilimumab (aCTLA-4/aPD-1) combination therapy has significantly improved clinical outcomes in patients with metastatic melanoma, with 50%-60% of patients responding to treatment, but predictors of response are poorly characterized. We hypothesized that circulating cytokines and peripheral white blood cells may predict response to therapy and evaluated 15 cytokines and complete blood counts (CBC with differentials) from 89 patients with advanced melanoma treated with combination therapy from three points in time: pre-treatment, one month and approximately three months after starting therapy. Clinical endpoints evaluated included durable clinical benefit (DCB), progression-free survival (PFS), and overall survival (OS). A parsimonious predictive model was developed to identify cytokines predictors of response to combination therapy. In this study, we found that pre-treatment, patients with DCB had higher IL-23, lower CXCL6, and lower IL-10 levels. Lower NLR one month after starting therapy predicted better PFS and OS, primarily driven by an increase in absolute lymphocytes. A multivariate model demonstrated that baseline CXCL6, IL-10, IL-23 were independent predictors of therapy response, and the combined model has reached an area under the curve (AUC) of 0.79 in prediction of response to combination therapy. Our study identified baseline CXCL6, IL-23, and IL-10 as predictors of response to aCTLA4/aPD1 combination therapy among patients with metastatic melanoma. This study also provides a framework for identifying patients who are likely to respond to combination ICB, as well as a subset of patients with high risk of developing resistance and are thus in need of alternative therapeutic options, such as clinical trials.
Collapse
Affiliation(s)
- Jiajia Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Giuseppe Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mariano Severgnini
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joanna Baginska
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason L Weirather
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Janice D Russell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy Y Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ryan Brennick
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew Nazzaro
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Emma Hathaway
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marta Holovatska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Claire Manuszak
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Srinika Ranasinghe
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
2
|
El-Ghazzi N, Monier A, Italiano A, Besson A, Angeli E. Immune-induced thrombocytopenia by pembrolizumab: case report and review of literature. Platelets 2025; 36:2487767. [PMID: 40178025 DOI: 10.1080/09537104.2025.2487767] [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: 07/25/2024] [Revised: 02/01/2025] [Accepted: 03/16/2025] [Indexed: 04/05/2025]
Abstract
Immune-checkpoint blockades (ICBs) are now used in early-stage diseases like triple-negative breast cancer (TNBC). While effective, they can cause severe toxicities. We report the first case of life-threatening immune thrombocytopenia (ITP) induced by pembrolizumab during neoadjuvant chemo-immunotherapy for early TNBC. A 42-year-old woman with early-stage TNBC developed grade 4 thrombocytopenia, diagnosed as ITP, after 107 days of pembrolizumab treatment. She required intensive care unit (ICU) admission and high-dose steroids, and intravenous immunoglobulin therapy, leading to a rapid recovery. ITP is a rare but potentially fatal complication of immunotherapy, with an incidence of less than 1% and a mortality rate of up to 20% in affected patients. Immediate recognition and steroid therapy are critical, as platelet transfusion is usually ineffective. Diagnosis is often delayed due to its similarity to chemotherapy-induced marrow toxicity. Immunotherapy-induced ITP generally contraindicates further use of the treatment. ITP, although uncommon, is a serious complication of immunotherapy requiring immediate intervention. The growing use of immunotherapy necessitates increased awareness of its potential toxicities among healthcare providers.
Collapse
Affiliation(s)
- Nathan El-Ghazzi
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Anna Monier
- Internal Medicine Department, Bordeaux University Hospital, Hôpital Pellegrin, University of Bordeaux, Bordeaux, France
| | - Antoine Italiano
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Aude Besson
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Eurydice Angeli
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| |
Collapse
|
3
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
4
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
5
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Robert C, Kicinski M, Dutriaux C, Routier É, Govaerts AS, Bührer E, Neidhardt EM, Durando X, Baroudjian B, Saiag P, Gaudy-Marqueste C, Ascierto PA, Arance A, Russillo M, Perrot JL, Mortier L, Aubin F, Dalle S, Grange F, Muñoz-Couselo E, Mary-Prey S, Amini-Adle M, Mansard S, Lebbe C, Funck-Brentano E, Monestier S, Eggermont AMM, Oppong F, Wijnen L, Schilling B, MandalÁ M, Lorigan P, van Akkooi ACJ. Combination of encorafenib and binimetinib followed by ipilimumab and nivolumab versus ipilimumab and nivolumab in patients with advanced melanoma with BRAF V600E or BRAF V600K mutations (EBIN): an international, open-label, randomised, controlled, phase 2 study. Lancet Oncol 2025; 26:781-794. [PMID: 40449497 DOI: 10.1016/s1470-2045(25)00133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Current first-line treatment for patients with metastatic melanoma with BRAFV600E or BRAFV600K mutations includes immunotherapy with immune checkpoint inhibitors and targeted therapy; however, the optimal sequencing of these treatments is unclear. We aimed to investigate the use of a targeted-therapy induction regimen before treatment with immune checkpoint inhibitors. METHODS This open-label, randomised, controlled, phase 2 trial (EBIN) was conducted at 37 centres in eight European countries. Eligible patients were 18 years or older and had previously untreated, unresectable, stage III or IV melanoma with BRAFV600E or BRAFV600K mutations and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) to one of two groups. Those in the induction group received targeted therapy (oral encorafenib 450 mg once a day plus oral binimetinib 45 mg twice a day for 12 weeks) followed by immune checkpoint inhibitors (intravenous nivolumab 3 mg/kg plus intravenous ipilimumab 1 mg/kg once every 3 weeks for four doses, followed by intravenous nivolumab 480 mg once every 4 weeks until unacceptable toxicity, disease progression, or 2 years of treatment). Patients in the control group received immune checkpoint inhibitors as above without any induction targeted therapy. Randomisation was conducted using a minimisation technique and was stratified by centre and a variable defined using stage and lactate dehydrogenase activity. The primary outcome was progression-free survival in the intention-to-treat population. Safety was assessed in all patients who initiated the protocol treatment. In this Article we report the primary analysis. The study is registered with ClinicalTrials.gov, NCT03235245, and is ongoing. FINDINGS Between Nov 12, 2018, and July 11, 2022, 271 patients were randomly assigned: 136 to the induction group and 135 to the control group. 103 (38%) patients were female, 168 (62%) were male, and the median age was 55 years (IQR 43-66). The median follow-up time was 21 months (IQR 13-33). There was no evidence of a longer progression-free survival in the induction group than in the control group (hazard ratio 0·87, 90% CI 0·67-1·12; p=0·36). The median progression-free survival was 9 months (95% CI 7-13) in the induction group and 9 months (5-14) in the control group. Grade 3-5 treatment-related adverse events occurred in 57 (42%) of 136 patients who started treatment in the induction group and in 42 (32%) of 131 patients who started treatment in the control group. The most common grade 3-4 treatment-related adverse event was hepatitis (17 [13%] of 136 patients in the induction group and nine [7%] of 131 patients in the control group). Serious treatment-related adverse events occurred in 45 (33%) of 136 patients in the induction group and 33 (25%) of 131 patients in the control group. There were three treatment-related deaths: two from cardiac events (heart failure and arrhythmia) in the induction group and one from meningitis in the control group. INTERPRETATION The targeted-therapy induction regimen did not improve progression-free survival compared with first-line treatment with immune checkpoint inhibitors in unselected patients with advanced melanoma with BRAFV600E or BRAFV600K mutations. FUNDING Bristol Myers Squibb and Pierre Fabre.
Collapse
Affiliation(s)
- Caroline Robert
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France.
| | | | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
| | - Émilie Routier
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, University of Paris-Saclay, Villejuif, France
| | | | | | | | - Xavier Durando
- INSERM U1240 IMoST, Université Clermont Auvergne, Clermont-Ferrand, France; Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France; Département d'Oncologie Médicale, Centre Jean Perrin, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR501, Clermont-Ferrand, France
| | - Barouyr Baroudjian
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Caroline Gaudy-Marqueste
- Dermatology and Skin Cancer Department, Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS 'Fondazione G Pascale', Naples, Italy
| | - Ana Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Michelangelo Russillo
- Sarcoma and Rare Tumours Departmental Unit, IRCCS Regina Elena National Cancer Institute Rome, Rome, Italy
| | - Jean-Luc Perrot
- Groupe d'Imagerie Cutanée Non Invasive (GICNI), Société Française de Dermatologie (SFD), Paris, France; Department of Dermatology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Laurent Mortier
- Department of Dermatology, INSERM U1189, CHU Lille, CARADERM, Lille University, Lille, France
| | - Francois Aubin
- Department of Dermatology, UHC and INSERM 1098, Besançon, France
| | - Stéphane Dalle
- Dermatology Department, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Florent Grange
- Dermatology/Oncology, CHU Reims-Hôpital Robert Debre, Reims, France; Dermatology Department, Valence Hospital, Valence, France
| | - Eva Muñoz-Couselo
- Department of Oncology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Sorilla Mary-Prey
- Service de Dermatologie, CHU de Bordeaux, Bordeaux, France; BRIC (Bordeaux Institute of Oncology), INSERM UMR1312, Université de Bordeaux, Bordeaux, France
| | | | - Sandrine Mansard
- Service de Dermatologie, Centre Hospitalo-Universitaire de Clermont Auvergne, Clermont-Ferrand, France
| | - Céleste Lebbe
- Université Paris Cité, AP-HP Dermato-oncology, Cancer Institute AP-HP, Nord Paris Cité, INSERM U976, Saint Louis Hospital, Paris, France
| | - Elisa Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise Paré Hospital, APHP & EA 4340 "Biomarkers in cancerology and hemato-oncology", UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Sandrine Monestier
- Dermatology and Skin Cancer Department Aix Marseille Univ, APHM, La Timone Hospital, Marseille, France
| | - Alexander M M Eggermont
- Board of Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximilians University, Munich, Germany; Princess Máxima Center, Utrecht, Netherlands
| | | | | | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany; Department of Dermatology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Mario MandalÁ
- Unit of Oncology, Santa Maria Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester and Christie NHS Foundation Trust, Manchester, UK
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
7
|
Day D, Ganju V, Chung K, Si L, Mao L, Aghmesheh M, Hoyer R, Brewin K, Zeng S, Zhang M, Lu Q, Jiang C, Ren F, Zhu Y, Guo J. First-in-human phase I study of EMB-02, a bispecific antibody targeting PD-1 and LAG-3 in patients with advanced solid tumors. Br J Cancer 2025; 132:905-912. [PMID: 40234667 DOI: 10.1038/s41416-025-02990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND EMB-02 is a symmetric bispecific antibody targeting programmed cell death protein-1 and lymphocyte-activation gene 3 simultaneously. Here, we present the first-in-human study results of EMB-02 in patients with advanced solid tumors. METHODS Patients were treated with intravenous infusions of EMB-02 at doses of 6-900 mg. The primary objective was to evaluate the safety and tolerability and to determine the maximum tolerated dose and/or recommended phase II dose(s). Secondary objectives included characterizing the pharmacokinetic (PK) profile, assessing preliminary antitumor activity and the immunogenicity. RESULTS A total of 47 patients were enrolled. All grade and grade 3/4 treatment-emergent and treatment related adverse events occurred in 97.9%, 48.9%, 68.1% and 12.8% patients, respectively. The objective response rate (ORR) was 6.4% and clinical benefit rate at 24 weeks (CBR-24) was 25.5% in overall population. The CBR-24 was 33.3% in checkpoint inhibitor (CPI)-naïve patients, and 15% in CPI-treated. No clear relationship was observed between the efficacy and PD-L1, LAG-3, or MHC II expression level. Doses 360 mg or higher resulted in sustained saturation of PD-1 receptors on circulating CD3 + T cells. CONCLUSIONS EMB-02 demonstrated a favorable safety profile and early efficacy signals in multiple solid tumors, warranting further development. (NCT04618393).
Collapse
Affiliation(s)
- Daphne Day
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Vinod Ganju
- Oncology Department, Peninsula And Southeast Oncology, Frankston, VIC, Australia
| | - Ki Chung
- Department of Medicine, Prisma Health Cancer Institute, Greenville, SC, USA
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Lili Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China
| | - Morteza Aghmesheh
- Medical Oncology Department, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Robert Hoyer
- Medical Oncology Department, UCHealth Memorial Hospital Central, Colorado Springs, CO, USA
| | - Kim Brewin
- Medical Oncology Department, Monash Health-Monash MedicalCentre, Clayton, VIC, Australia
| | - Shuqi Zeng
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Mingfei Zhang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Qiaoyang Lu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Chengjun Jiang
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Fang Ren
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Yonghong Zhu
- Clinical Development, Shanghai EpimAb Biotherapeutics Co., Ltd., Shanghai, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Collaborative Innovation Center for Cancer Medicine, Peking UniversityCancer Hospital and Institute, Beijing, China.
| |
Collapse
|
8
|
Broche C, Um K, Vilgrain I, Mouret S, Leccia MT, Seigneurin A, Bouillet L, Enquebecq M, Salomon A, Aspord C, Busser B, Porret P, Belbezier A, Chaperot L, Charles J. Soluble VE-cadherin as a biomarker of response to immunotherapy by anti-PD1 in metastatic melanoma. Br J Cancer 2025; 132:863-865. [PMID: 40251278 DOI: 10.1038/s41416-025-03026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Clémentine Broche
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Khémary Um
- Institute for Advanced Biosciences Team Immunobiology and Immunotherapy in Chronic Diseases, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Grenoble, France
- Etablissement français du sang, Research and Development Laboratory, Grenoble, France
| | - Isabelle Vilgrain
- Biomicrotechnology and Functional Genomics (Biomics), Biosciences and Bioengineering for Health Laboratory, Univ. Grenoble Alpes, CNRS, InsermU 13, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Grenoble, France
| | - Stéphane Mouret
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
- Team Cancer Targets and Experimental Therapeutics, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Marie-Thérèse Leccia
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences Team Immunobiology and Immunotherapy in Chronic Diseases, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Grenoble, France
| | - Arnaud Seigneurin
- Laboratory TIMC, Univ. Grenoble Alpes, CNRS, Grenoble, France
- Medical evaluation Unit, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Laurence Bouillet
- Department of Internal Medicine, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Marie Enquebecq
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Aude Salomon
- Biology and Biotechnology for Health Laboratory, Univ. Grenoble Alpes, CNRS, InsermU 1292, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences Team Immunobiology and Immunotherapy in Chronic Diseases, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Grenoble, France
- Etablissement français du sang, Research and Development Laboratory, Grenoble, France
| | - Benoit Busser
- Team Cancer Targets and Experimental Therapeutics, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Institute for Advanced Biosciences, Grenoble, France
- Department of Clinical Biochemistry, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Pauline Porret
- Department of Radiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
| | - Aude Belbezier
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France.
- Team Cancer Targets and Experimental Therapeutics, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Institute for Advanced Biosciences, Grenoble, France.
| | - Laurence Chaperot
- Institute for Advanced Biosciences Team Immunobiology and Immunotherapy in Chronic Diseases, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Grenoble, France
- Etablissement français du sang, Research and Development Laboratory, Grenoble, France
| | - Julie Charles
- Department of Dermatology, Allergology and Photobiology, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences Team Immunobiology and Immunotherapy in Chronic Diseases, Univ. Grenoble Alpes, CNRS UMR 5309, Inserm U 1209, Grenoble, France
| |
Collapse
|
9
|
Amrane K, Le Noac'h P, Hemon P, Abgral R, Le Meur C, Pradier O, Misery L, Legoupil D, Berthou C, Uguen A. MHC class II: a predictor of outcome in melanoma treated with immune checkpoint inhibitors. Melanoma Res 2025; 35:176-186. [PMID: 39945603 DOI: 10.1097/cmr.0000000000001022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
This study aimed to evaluate the predictive value of MHC class II (MHC-II) expression by melanoma cells in a large cohort of metastatic cutaneous melanoma patients treated with immune checkpoint inhibitors (ICIs). We conducted a single-center, retrospective study involving stage IV cutaneous melanoma patients who received ICI as first-line therapy. MHC-II expression in melanoma cells was quantified using dual-color anti-SOX10 and anti-MHC-II immunohistochemistry on tumor samples from 95 patients. The primary endpoint was event-free survival (EFS), with secondary endpoints including 1-year EFS, 1-year overall survival (OS), disease control rate (DCR), and the correlation between MHC-II expression and clinico-biological characteristics. The cohort had a median age of 67 years (range, 33-90), with a male-to-female ratio of 50 : 45. Thirty-three percent of patients received the ipilimumab-nivolumab combination. The median follow-up was 16.8 months. Disease progression occurred in 58 patients (61%), with a median time to progression of 4.8 months. Forty-six patients (48.4%) experienced an event within the first year, and 52 patients (54.7%) died during follow-up. MHC-II positivity was observed in ≥10% of melanoma cells in 6.3% of patients. MHC-II expression was significantly associated with 1-year EFS ( P = 0.037) and DCR ( P = 0.032), but not with EFS or 1-year OS. Age, phototype, and brain metastases were correlated with MHC-II expression status. Our findings suggest that MHC-II expression by melanoma cells may serve as a favorable predictive biomarker for survival in metastatic cutaneous melanoma patients treated with ICIs.
Collapse
Affiliation(s)
- Karim Amrane
- Department of Oncology, Regional Hospital of Morlaix, Morlaix
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Pierre Le Noac'h
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Pathology, University Hospital of Brest
| | - Patrice Hemon
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital of Brest
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| | - Coline Le Meur
- Department of Radiotherapy, University Hospital of Brest
| | - Olivier Pradier
- Department of Radiotherapy, University Hospital of Brest
- Inserm, UMR1101, LaTIM, University of Western Brittany
| | - Laurent Misery
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Delphine Legoupil
- Department of Dermatology, University Hospital of Brest
- Laboratoire sur les Interactions Épithéliums-Neurones (LIEN-EA4685), Université de Bretagne Occidentale
| | - Christian Berthou
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- Department of Hematology, University Hospital of Brest, Brest, France
| | - Arnaud Uguen
- Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO
- UMR Inserm 1304 GETBO, IFR 148, University of Western Brittany
| |
Collapse
|
10
|
Sperduto PW, Marqueen KE, Chang E, Li J, Davies MA, Ebner DK, Breen WG, Lamba N, Shih HA, Edwards D, Kim MM, Mahal A, Rahman R, Ankrah N, Boggs DH, Lewis C, Hyer D, Buatti JM, Johri F, Soliman H, Masucci L, Roberge D, Aneja S, Chiang V, Phuong C, Braunstein S, Dajani S, Sachdev S, Wan Z, Niedzwiecki D, Vaios E, Kirkpatrick JP, Pasetsky J, Wang TJ, Kutuk T, Kotecha R, Ross RB, Rusthoven CG, Nakano T, Tawbi HA, Mehta MP. Improved Survival and Prognostication in Melanoma Patients With Brain Metastases: An Update of the Melanoma Graded Prognostic Assessment. J Clin Oncol 2025; 43:1910-1919. [PMID: 40245362 PMCID: PMC12119226 DOI: 10.1200/jco-24-01351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/24/2025] [Accepted: 02/21/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Survival for patients with melanoma has recently improved. The propensity of melanoma to metastasize to the brain remains a common and serious feature of this disease. The purposes of this study were to evaluate prognostic factors for patients with newly diagnosed melanoma brain metastases (MBMs) in a large cohort treated with modern multimodal therapies, compare those results with those in prior eras, and update the Melanoma Graded Prognostic Assessment (GPA). METHODS Univariable and multivariable (MVA) analyses of prognostic factors and treatments associated with survival were performed on 1,796 patients with newly diagnosed MBM treated between January 01, 2015, and December 31, 2021, using a multi-institutional retrospective database. Multiple imputation was used to address missingness of potential predictors. Significant variables in combined MVA were used to update the Melanoma GPA. Comparisons were made with legacy cohorts. RESULTS Median survivals for cohorts A (1985-2007, n = 481), B (2006-2015, n = 823), and C (2015-2021, n = 1,796) were 6.7, 9.8, and 16.6 months and median follow-up times were 40.1, 43.6, and 48.8 months, respectively. In combined MVA, significant prognostic factors for survival were higher Karnofsky Performance Status, fewer MBMs, absence of extracranial metastases, lower serum lactate dehydrogenase, and no immunotherapy before MBM. These factors were incorporated into the updated Melanoma GPA. The combined median and 3-year survivals for patients with GPA 0-1, 1.5-2, and 2.5-4.0 were 5.4, 13.2, and 43.2 months and 12.4%, 28.8%, and 51.6%, respectively. CONCLUSION Prognostic factors have changed and survival has improved for patients with MBM but varies widely by GPA. The updated Melanoma GPA calculator (BrainMetGPA), available free online, can be used to estimate survival, individualize treatment, stratify clinical trials, guide surveillance, and augment clinical trial eligibility. Multidisciplinary treatment is essential. Trials are needed to elucidate the optimal sequencing of various therapeutic modalities.
Collapse
Affiliation(s)
| | | | - Enoch Chang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Li
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | - Nii Ankrah
- University of Alabama, Birmingham, Birmingham, AL
| | | | | | | | | | - Fasila Johri
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Hany Soliman
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, Canada
| | - Laura Masucci
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | - David Roberge
- Centre Hospitalier de l’Universite de Montreal, Montreal, Canada
| | | | | | | | | | | | | | - Zihan Wan
- Duke Cancer Institute-Biostatistics Shared Resource, Durham, NC
| | | | | | | | | | | | - Tugce Kutuk
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Rupesh Kotecha
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | | | | | | | | | - Minesh P. Mehta
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
11
|
Güven DC, Thong MS, Arndt V. Survivorship outcomes in patients treated with immune checkpoint inhibitors: a scoping review. J Cancer Surviv 2025; 19:806-845. [PMID: 38175366 PMCID: PMC12081552 DOI: 10.1007/s11764-023-01507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have become a central part of cancer care. However, the survivorship outcomes in patients treated with ICIs are understudied. Therefore, we conducted a scoping review to evaluate the current status of the field and to establish research gaps regarding survivorship outcomes with ICIs in real-life cohorts. METHODS We used the Web of Science, PubMed, and Embase databases to systematically filter published studies with real-life cohorts from January 1, 2010, until October 19, 2022. Studies evaluating at least one survivorship outcome in ICI-treated patients were included. RESULTS A total of 39 papers were included. Quality of life (QoL) (n = 23), toxicity burden (n = 16), and psychosocial issues (n = 9) were the most frequently evaluated survivorship outcomes. Anti-PD-1/PD-L1 monotherapy and a response to treatment were associated with better QoL. In addition, the ICIs were associated with grade 3 or higher immune-related adverse events (irAEs) in 10-15% and late/long-term irAEs in 20-30% of the survivors. Regarding psychosocial problems, over 30% of survivors showed evidence of anxiety and depression, and 30-40% of survivors reported neurocognitive impairments. CONCLUSION The survivors treated with ICIs have impairments in most survivorship domains. Further research is needed to gather data on the understudied survivorship outcomes like late and long-term effects, fertility, financial toxicity, and return to work in survivors treated with ICIs. IMPLICATIONS FOR CANCER SURVIVORS Available evidence demonstrates that a significant portion of survivors treated with ICIs have a significant toxicity burden, lower QoL than the general population, and a high rate of psychosocial problems.
Collapse
Affiliation(s)
- Deniz Can Güven
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Sihhiye, Ankara, Turkey.
- Health Sciences University, Elazig City Hospital, Elazig, Turkey.
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Melissa Sy Thong
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Arndt
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
Olsson-Brown A, Jain A, Frazer R, Farrugia D, Carser J, Houghton J, Lewis RD, D'Mello S, Emanuel G. Clinical Management and Outcomes of Immune-Related Adverse Events During Treatment with Immune Checkpoint Inhibitor Therapies in Melanoma and Renal Cell Carcinoma: A UK Real-World Evidence Study. Oncol Ther 2025:10.1007/s40487-025-00349-z. [PMID: 40448748 DOI: 10.1007/s40487-025-00349-z] [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: 02/07/2025] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
INTRODUCTION Immune checkpoint inhibitor (ICI) therapy, although effective in treating patients with a variety of advanced malignancies, can result in potentially severe or even fatal immune-related adverse events (irAEs). This study aimed to generate real-world evidence on irAE characteristics, clinical management and clinical outcomes among patients with advanced (unresectable or metastatic) malignant melanoma (a/mMM) or advanced renal cell carcinoma (aRCC) treated with nivolumab (NIVO) and/or ipilimumab (IPI) in the UK. METHODS This was a multi-centre, retrospective cohort study of adult patients with a/mMM or aRCC, who received NIVO and/or IPI at one of five specialist treatment centres in the UK between 1 January 2016 and 31 March 2020. The incidence and grading of irAEs were described, as well as time to irAE onset, the management of irAEs and overall survival (OS). RESULTS In total, 199 patients were included in the study: 162 with a/mMM and 37 with aRCC. The majority of patients in both a/mMM (75.3%) and aRCC (62.2%) cohorts reported any irAE, while 45.9% and 30.4% in the a/mMM and aRCC cohorts reported grade 3 or 4 irAEs, respectively. Colitis/diarrhoea, skin reactions and hepatitis were most frequently reported, and the predominant treatment prescribed for any irAE was corticosteroids only. Analysis indicated a positive association between the development of an irAE and longer OS. CONCLUSIONS Findings from this study support current literature, provide further insights into the characteristics and clinical management of irAEs and support an association between the development of an irAE and improved OS in these patient groups.
Collapse
Affiliation(s)
| | - Ankit Jain
- The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | - David Farrugia
- Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | | | - John Houghton
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Ruth D Lewis
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Simon D'Mello
- Bristol Myers Squibb, Sanderson Road, Denham, Uxbridge, UB8 1DH, UK.
| | | |
Collapse
|
13
|
Miglino N, Toussaint NC, Ring A, Bonilla X, Tusup M, Gosztonyi B, Mehra T, Gut G, Jacob F, Chevrier S, Lehmann KV, Casanova R, Jacobs A, Sivapatham S, Boos L, Rahimzadeh P, Schuerch M, Sobottka B, Chicherova N, Yu S, Wegmann R, Mena J, Milani ES, Goetze S, Esposito C, Sarabia Del Castillo J, Frei AL, Nowak M, Irmisch A, Kuipers J, Baciu-Drăgan MA, Ferreira PF, Singer F, Bertolini A, Prummer M, Lischetti U, Tumor Profiler Consortium, Aebersold R, Bacac M, Maass G, Moch H, Weller M, Theocharides APA, Manz MG, Beerenwinkel N, Beisel C, Pelkmans L, Snijder B, Wollscheid B, Heinzelmann V, Bodenmiller B, Levesque MP, Koelzer VH, Rätsch G, Dummer R, Wicki A. Feasibility of multiomics tumor profiling for guiding treatment of melanoma. Nat Med 2025:10.1038/s41591-025-03715-6. [PMID: 40425842 DOI: 10.1038/s41591-025-03715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 04/09/2025] [Indexed: 05/29/2025]
Abstract
There is limited evidence supporting the feasibility of using omics and functional technologies to inform treatment decisions. Here we present results from a cohort of 116 melanoma patients in the prospective, multicentric observational Tumor Profiler (TuPro) precision oncology project. Nine independent technologies, mostly at single-cell level, were used to analyze 126 patient samples, generating up to 500 Gb of data per sample (40,000 potential markers) within 4 weeks. Among established and experimental markers, the molecular tumor board selected 54 to inform its treatment recommendations. In 75% of cases, TuPro-based data were judged to be useful in informing recommendations. Patients received either standard of care (SOC) treatments or highly individualized, polybiomarker-driven treatments (beyond SOC). The objective response rate in difficult-to-treat palliative, beyond SOC patients (n = 37) was 38%, with a disease control rate of 54%. Progression-free survival of patients with TuPro-informed therapy decisions was 6.04 months, (95% confidence interval, 3.75-12.06) and 5.35 months (95% confidence interval, 2.89-12.06) in ≥third therapy lines. The proof-of-concept TuPro project demonstrated the feasibility and relevance of omics-based tumor profiling to support data-guided clinical decision-making. ClinicalTrials.gov identifier: NCT06463509 .
Collapse
Affiliation(s)
- Nicola Miglino
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Swiss Data Science Center SDSC, Zurich, Switzerland
| | - Alexander Ring
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Ximena Bonilla
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
| | - Marina Tusup
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Benedict Gosztonyi
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Tarun Mehra
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Gabriele Gut
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Stephane Chevrier
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Kjong-Van Lehmann
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
- Department of Biology, RWTH Aachen, Aachen, Germany
| | - Ruben Casanova
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Andrea Jacobs
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Sujana Sivapatham
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Laura Boos
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Parisa Rahimzadeh
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Manuel Schuerch
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Bettina Sobottka
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Natalia Chicherova
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Shuqing Yu
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Swiss Data Science Center SDSC, Zurich, Switzerland
| | - Rebekka Wegmann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Julien Mena
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Emanuela S Milani
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), ETH Zurich, Zurich, Switzerland
| | - Cinzia Esposito
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Anja L Frei
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Anja Irmisch
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Jack Kuipers
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Monica-Andreea Baciu-Drăgan
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Pedro F Ferreira
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Franziska Singer
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Anne Bertolini
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Michael Prummer
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ulrike Lischetti
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Rudolf Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Gerd Maass
- Roche Diagnostics GmbH, MWG, Penzberg, Germany
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Berend Snijder
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Viola Heinzelmann
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital, Zurich, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Gunnar Rätsch
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Computer Science, Institute of Machine Learning, ETH Zurich, Zurich, Switzerland
- Biomedical Informatics, University Hospital Zurich, Zurich, Switzerland
- AI Center at ETH Zurich, ETH Zurich, Zurich, Switzerland
- Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital, Zurich, Switzerland.
| |
Collapse
Collaborators
Melike Ak, Faisal S Al-Quaddoomi, Silvana I Albert, Jonas Albinus, Ilaria Alborelli, Sonali Andani, Per-Olof Attinger, Monica-Andreea Baciu-Drăgan, Daniel Baumhoer, Beatrice Beck-Schimmer, Lara Bernasconi, Lars Bosshard, Byron Calgua, Stéphane Chevrier, Ricardo Coelho, Maya D'Costa, Esther Danenberg, Natalie R Davidson, Stefanie Engler, Martin Erkens, Katja Eschbach, André Fedier, Joanna Ficek-Pascual, Bruno Frey, Linda Grob, Detlef Günther, Pirmin Haeuptle, Viola Heinzelmann-Schwarz, Sylvia Herter, Rene Holtackers, Tamara Huesser, Alexander Immer, Tim M Jaeger, Alva R James, Philip M Jermann, André Kahles, Abdullah Kahraman, Werner Kuebler, Christian P Kunze, Christian Kurzeder, Mitchell Levesque, Flavio C Lombardo, Sebastian Lugert, Philipp Markolin, Martin Mehnert, Julian M Metzler, Simone Muenst, Riccardo Murri, Charlotte K Y Ng, Stefan Nicolet, Monica Nunez Lopéz, Patrick Ga Pedrioli, Salvatore Piscuoglio, Laurie Prélot, Natalie Rimmer, Mathilde Ritter, Christian Rommel, María L Rosano-González, Natascha Santacroce, Ramona Schlenker, Petra C Schwalie, Severin Schwan, Tobias Schär, Gabriela Senti, Wenguang Shao, Vipin T Sreedharan, Stefan Stark, Daniel J Stekhoven, Tanmay Tanna, Tinu M Thomas, Markus Tolnay, Vinko Tosevski, Mustafa A Tuncel, Audrey Van Drogen, Marcus Vetter, Tatjana Vlajnic, Sandra Weber, Walter P Weber, Fabian Wendt, Norbert Wey, Mattheus He Wildschut, Johanna Ziegler, Marc Zimmermann, Martin Zoche, Gregor Zuend,
Collapse
|
14
|
Papayanis PN, Au C, Jelinek M, Tan A. Using liquid biopsies to guide treatment and monitor response in BRAF V600E positive adenocarcinoma of unknown primary. BMJ Case Rep 2025; 18:e264469. [PMID: 40425210 PMCID: PMC12107294 DOI: 10.1136/bcr-2024-264469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Liquid biopsies using circulating tumour DNA (ctDNA) have emerged as an alternative to conventional biopsies. They can be used to aid in diagnosing and selecting an agent for treatment and can possibly be used to monitor disease response to treatment. In this report, we present a patient who initially presented with lower abdominal pain. Imaging showed extensive retroperitoneal lymphadenopathy and lymph node biopsy demonstrated poorly differentiated carcinoma. Further workup did not reveal a primary lesion, but his genetic analysis revealed a BRAF V600E mutation and CD274 amplification which was used to guide treatment of the adenocarcinoma as a melanoma of unknown primary. He was initiated on ipilimumab and nivolumab and his ctDNA levels showed rapid improvement. After treatment was stopped due to adverse events, he was monitored via ctDNA, with an increase prompting repeat imaging that demonstrated enlargement of his lesions prompting a resumption of treatment.
Collapse
Affiliation(s)
| | - Cherry Au
- Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Michael Jelinek
- Hematology, Oncology and Cellular Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Alan Tan
- Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
15
|
Noringriis IM, Donia M, Madsen K, Schmidt H, Haslund CA, Bastholt L, Svane IM, Ellebaek E. Long-term clinical outcome of patients with metastatic melanoma and initial stable disease during anti-PD-1 checkpoint inhibitor immunotherapy with pembrolizumab. Br J Cancer 2025:10.1038/s41416-025-03048-8. [PMID: 40419744 DOI: 10.1038/s41416-025-03048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND A substantial number of patients with metastatic melanoma (MM) treated with anti-PD-1 monotherapy have initial stable disease (SD), yet the real-world prognosis of these patients remains unclear. METHODS In this nationwide cohort study, we analysed real-world outcomes of patients with MM treated with pembrolizumab in Denmark. Focusing on patients with initial SD, we assessed best overall response (BOR), progression-free survival (PFS), and overall survival (OS) and identified predictors of survival in multivariable analyses. RESULTS Out of 1048 included patients, 233 (22.2%) had initial SD with a median PFS and OS of 14.7 and 50.1 months. Subsequent partial response (PR) or complete response (CR) was developed by 44 (18.9%) and 52 (22.3%) patients showing significantly improved PFS compared to patients with continued SD (PR: HR 0.52, 95% CI 0.34-0.81, p = 0.003; CR: HR 0.15, 95% CI 0.07-0.32, p < 0.001) and survival rates comparable to patients with initial PR and CR, respectively. Furthermore, 49 (21.0%) patients showed continued disease control (median follow-up of 82.3 months). For 51.0% of these patients, the last dose of pembrolizumab was administered during SD with a median treatment duration of 12.4 months. CONCLUSIONS Of patients with initial SD, 40% developed a subsequent objective response with improved long-term prognosis comparable to patients with initial response. More than 20% exhibited continued disease control.
Collapse
Affiliation(s)
- Inge Mansfield Noringriis
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Kasper Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| |
Collapse
|
16
|
Biteau M, Sibaud V, Maria A, Dion J, Uro-Coste E, Siegfried A, Pastissier A, Cougoul P, Delavigne K, Comont T, Rivet V. Immune checkpoint inhibitor-related eosinophilic fasciitis: 3 case reports with literature review. Rev Med Interne 2025:S0248-8663(25)00614-9. [PMID: 40399177 DOI: 10.1016/j.revmed.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are associated with a large spectrum of immune-related Adverse Events (irAEs). ICIs can exceptionally induce eosinophilic fasciitis (EF). Data of ICI-related EF (ICI-EF) are scarce without clear guidelines on both diagnosis and treatment. METHODS We conducted a literature review of all cases of patients over 18 years old with EF diagnosis following ICI treatment between 2016 and November 2023 and meeting Pinal-Fernandez's criteria. New cases from two departments (French Region of Occitania) were added to complete the cohort. RESULTS We selected 30 cases: 27 from literature review and 3 new cases diagnosed in Occitania. There was male predominance, with a sex ratio (M/F) of 1.3 with a median age of 57 years. The most common tumor was melanoma (50%). The time to onset was 10 months. Twenty-two patients (73%) had hypereosinophilia (mean 3300/mm3). A large majority (25/30, 83%) received at least one course of steroids therapy while immunosuppressive treatment was needed for 18 patients (72%). Rechallenge was proposed in 2 patients without recurrence of EF. DISCUSSION Clinical and biological characteristics appeared roughly similar between ICI-EF and idiopathic-EF. Management is currently based on clinical practice of idiopathic-EF with corticosteroids often used in first line of treatment. However, a second line (methotrexate of mycophenolate mofetil) was required in majority of patients as observed in idiopathic-EF. CONCLUSION There is a significant reporting signal of EF following ICIs. A proactive management is required in all cases. A specific algorithm for treatment is finally proposed.
Collapse
Affiliation(s)
- Mélanie Biteau
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France
| | - Vincent Sibaud
- Oncoderrmatology Department - Medical Oncology, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, Toulouse, France
| | - Alexandre Maria
- Internal Medicine and Immuno-Oncology Department (MEDI(2)O), Hospital Saint Eloi, Montpellier University Hospital, Montpellier, France
| | - Jérémie Dion
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France; Internal Medicine and Immuno-Oncology Department (MEDI(2)O), Hospital Saint Eloi, Montpellier University Hospital, Montpellier, France
| | - Emanuelle Uro-Coste
- Pathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, Toulouse, France
| | - Aurore Siegfried
- Pathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, Toulouse, France
| | - Andréa Pastissier
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France
| | - Pierre Cougoul
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France
| | - Karen Delavigne
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France
| | - Tthibault Comont
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France
| | - Valérian Rivet
- Internal Medicine and Immunopathology Department, The Cancer University of Toulouse Oncopole, Toulouse University Hospital, 1, avenue Joliot-Curie, 31100 Toulouse, France.
| |
Collapse
|
17
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Nardin C, Vernerey D, Aubin F. Ipilimumab + nivolumab in combination vs. nivolumab monotherapy as first-line treatment in patients with metastatic melanoma: conclusions from a real-world French national cohort. Br J Dermatol 2025; 192:971-972. [PMID: 40036378 DOI: 10.1093/bjd/ljaf080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Affiliation(s)
- Charlée Nardin
- Hôpital Jean Minjoz, Besançon, Bourgogne-Franche-Comté, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, Besançon University Hospital Center, Besançon, Bourgogne-Franche-Comté, France
| | - François Aubin
- Methodology and Quality of Life Unit in Oncology, Besançon University Hospital Center, Besançon, Bourgogne-Franche-Comté, France
| |
Collapse
|
19
|
Luke JJ, Bever K, Hodi FS, Taube J, Massey A, Yao D, Neely J, Tam R, Lee G, Gupta A, Dutta S, Szabo P, Bao R, Reilly T. Rationale and feasibility of a rapid integral biomarker program that informs immune-oncology clinical trials: the ADVISE trial. J Immunother Cancer 2025; 13:e011170. [PMID: 40389374 PMCID: PMC12090868 DOI: 10.1136/jitc-2024-011170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND ADVISE (ADaptiVe biomarker trial that InformS Evolution of therapy) (NCT03335540) was a biomarker-adapted feasibility clinical trial of immunohistochemistry (IHC) to inform combination immuno-oncology (I-O) treatment. METHODS To inform I-O combination selection, messenger RNA expression analyses from The Cancer Genome Atlas evaluated associations between programmed death 1/programmed death ligand 1 (PD-1/PD-L1) and other I-O-associated genes. Tumor tissue blocks of melanoma, non-small cell lung cancer, renal cell carcinoma, urothelial carcinoma, squamous cell carcinoma of the head and neck, and gastroesophageal junction/gastric cancer were stained by IHC to assess expression of CD8, colony-stimulating factor 1 receptor, glucocorticoid-induced tumor necrosis factor receptor (GITR), indoleamine 2,3-dioxygenase 1, lymphocyte-activation gene 3, NKp46, forkhead box P3, and PD-L1. These results facilitated an I-O treatment selection algorithm where patient biopsy results dictated allocation into combinations of nivolumab with cabiralizumab, urelumab, linrodostat mesylate, relatlimab, BMS-986156 (anti-GITR), lirilumab, ipilimumab, or irradiation. The primary endpoint was the proportion of patients with qualified baseline tumor biopsy specimens where decision-enabling biomarker analysis was completed within 12 business days to select an I-O combination therapy. RESULTS Correlation of PD-1/L1 and I-O-associated genes varied across the spectrum of T-cell-inflamed versus non-inflamed tumors; however, tumors with low/intermediate PD-L1 expression demonstrated distinct upregulation of immune markers grouped by cell type (T cell, macrophage, etc). IHC analyses of I-O naïve tumors corroborated these findings with distinct immune target upregulation in low-to-intermediate inflamed tumors and significant associations between IHC-detected markers and T-cell inflammation score across most markers. In the clinical trial, 20/23 (87%) of eligible patients were successfully allocated and started on treatment within the 12-day window, meeting the primary endpoint. The safety profile appeared to generally align with those reported for the individual combinations from other trials. No treatment responses occurred. Most patients were allocated to the cabiralizumab treatment arm. CONCLUSIONS Actualization of a patient-specific I-O combination treatment selection strategy is feasible, however, determination of de novo integral biomarker thresholds of novel I-O targets to facilitate effective treatment of PD-1-refractory cancer remains fraught. These data emphasize the difficulty of integral biomarker development for I-O in translating from immunotherapy treatment-naïve biospecimens to the selection of patients in the PD-1-refractory state. TRIAL REGISTRATION NUMBER NCT03335540.
Collapse
Affiliation(s)
| | | | - F Stephen Hodi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | - David Yao
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jaclyn Neely
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Rachel Tam
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - George Lee
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | | | - Peter Szabo
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Tim Reilly
- Bristol Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
20
|
Billard K, Mortier L, Dereure O, Dalac S, Montaudié H, Legoupil D, Dutriaux C, De Quatrebarbes J, Maubec E, Leccia MT, Granel-Brocard F, Brunet-Possenti F, Arnault JP, Gaudy-Marqueste C, Pages C, Saiag P, L'Orphelin JM, Zehou O, Lesimple T, Allayous C, Porcher R, Oriano B, Dalle S, Lebbé C. The efficacy and safety of first-line metastatic melanoma treatment with ipilimumab + nivolumab vs. nivolumab in a real-world setting. Br J Dermatol 2025; 192:1096-1105. [PMID: 39605282 DOI: 10.1093/bjd/ljae470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/21/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND The Checkmate 067 randomized controlled trial, published in 2015, demonstrated improved progression-free survival (PFS) and numerically, although not statistically, superior overall survival (OS) for ipilimumab + nivolumab (I + N). OBJECTIVES The objective of this study was to compare the efficacy and safety of N with I + N as first-line treatment for metastatic melanoma in a real-world setting. METHODS Patients were prospectively included in the French MelBase cohort from 2013 to 2022. Eligible patients were those in first-line treatment for stage IIIc or IV melanoma, undergoing immunotherapy with N or I + N. The primary endpoint was OS at 36 months. The secondary endpoints included PFS at 36 months, best radiological response, and safety analyses. We conducted a propensity score using the inverse probability of treatment weighting (IPTW) method to overcome the various confounding factors and also a subgroup analysis (brain metastasis, lactate dehydrogenase levels and BRAF mutation status). RESULTS Patients were treated with N (n = 406) or I + N (n = 416). OS at 36 months was higher in the I + N group at 57.1% [95% confidence interval (CI) 50.7-64.2] than in the N group [46.6% (95% CI 41.6-52.1)]; hazard ratio (HR) 1.4 (95% CI 1.1-1.8). PFS at 36 months was significantly improved in the I + N group (42.3%) compared with the N group (21.9%), with a HR of 1.6 (95% CI 1.4-1.9). The objective response rate (ORR) was similar for the two groups (44%). The overall incidence of side-effects was comparable (82% vs. 84%), and severe toxicity (grade ≥ 3) was more frequent, although not significantly so, in the I + N arm vs. the N arm (41% vs. 29%). CONCLUSIONS Our results are consistent with those from the Checkmate 067 study, except for the ORR and the incidence of toxicities, which proved to be lower in our analysis.
Collapse
Affiliation(s)
- Karine Billard
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Clara Allayous
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | - Bastien Oriano
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| | | | - Céleste Lebbé
- Service d'onco-dermatologie, Hôpital Saint Louis APHP, Paris, France
| |
Collapse
|
21
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
22
|
Giri VK, McDermott DF, Zaemes J. The emerging role of lymphocyte-activation gene 3 targeting in the treatment of solid malignancies. Cancer 2025; 131:e35892. [PMID: 40344213 DOI: 10.1002/cncr.35892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
PD-(L)1-based immune checkpoint inhibitor therapies have profoundly impacted the treatment of many solid malignancies. Although the addition of CTLA-4 checkpoint inhibitors can enhance anticancer activity, it also significantly increases the rate of immune-related adverse events. Therefore, there has been much interest in identifying additional immune checkpoints to improve the outcomes seen with PD-1-based therapy while minimizing additional side effects. One such target, lymphocyte-activation gene 3 (LAG-3), has long been recognized as an important inhibitor of T-cell function via modulation of the T-cell receptor pathway. Several drugs targeting LAG-3 have been developed, including most prominently the monoclonal antibody relatlimab. To date, the most significant demonstration of efficacy in targeting LAG-3 has been the use of relatlimab with the PD-1 inhibitor nivolumab in the treatment of advanced melanoma. The combination of nivolumab plus relatlimab is more efficacious compared to PD-1 inhibition alone, as has been previously seen with the combination of CTLA-4 inhibitor ipilimumab with nivolumab. However, nivolumab plus relatlimab offers a potentially more favorable toxicity profile. Here, the authors review the mechanism of the LAG-3 pathway and its rationale as a target for anticancer therapy as well as currently available data regarding the use of LAG-3 agents in treating melanoma and other solid tumors. Other investigational agents that target LAG-3 via novel mechanisms are also reviewed.
Collapse
Affiliation(s)
- Vinay K Giri
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David F McDermott
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jacob Zaemes
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Barroso-Sousa R, Zanudo JGT, Li T, Reddy SM, Emens LA, Kuntz TM, Silva CAC, AlDubayan SH, Chu H, Overmoyer B, Lange P, DiLullo MK, Montesion M, Kasparian J, Hughes ME, Attaya V, Basta A, Lin NU, Tayob N, Jeselsohn R, Mittendorf EA, Tolaney SM. Nivolumab plus low-dose ipilimumab in hypermutated HER2-negative metastatic breast cancer: a phase II trial (NIMBUS). Nat Commun 2025; 16:4430. [PMID: 40360544 PMCID: PMC12075640 DOI: 10.1038/s41467-025-59695-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
In the phase II NIMBUS trial, patients with human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC) and high tumor mutational burden (TMB ≥ 9 mut/Mb) received nivolumab (3 mg/kg biweekly) and low-dose ipilimumab (1 mg/kg every 6 weeks) for 2 years or until progression. The primary endpoint was objective response rate (ORR) per RECIST 1.1 criteria. Among 30 patients enrolled, the median TMB was 10.9 mut/Mb (range: 9-110) and the confirmed objective response rate was 20%. Secondary endpoints included progression-free survival, overall survival, clinical benefit rate, and safety and tolerability, including immune-related adverse events (irAEs). A prespecified correlative outcome was to evaluate the ORR in patients with a TMB ≥ 14 mut/Mb. Patients with TMB ≥ 14 mut/Mb (n = 6) experienced higher response rates (60% vs 12%; p = 0.041) and showed a trend towards improved progression-free survival and overall survival compared to patients with TMB < 14 mut/Mb. Exploratory genomic analyses suggested that ESR1 and PTEN mutations may be associated with poor response, while clinical benefit was associated with a decrease or no change in tumor fraction by serial circulating tumor DNA during treatment. Stool microbiome analysis revealed that baseline blood TMB, PD-L1 positivity, and immune-related diarrhea are associated with distinct taxonomic profiles. In summary, some patients with hypermutated HER2-negative MBC experience extended clinical benefit with a dual immunotherapy regimen; a higher TMB, and additional genomic and microbiome biomarkers may optimize patient selection for therapy with nivolumab plus low-dose ipilimumab. (Funded by Bristol Myers Squibb; ClinicalTrials.gov identifier, NCT03789110).
Collapse
Affiliation(s)
| | - Jorge Gomez Tejeda Zanudo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tianyu Li
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Leisha A Emens
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Thomas M Kuntz
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Hoyin Chu
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Beth Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paulina Lange
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Molly K DiLullo
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | | | - Julie Kasparian
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Melissa E Hughes
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Victoria Attaya
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Ameer Basta
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Nancy U Lin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nabihah Tayob
- Harvard Medical School, Boston, MA, USA
- Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rinath Jeselsohn
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Zhou Z, Yang Y, Chen S, You M. Cost-effectiveness analysis of first-line cadonilimab plus chemotherapy in HER2-negative advanced gastric or gastroesophageal junction adenocarcinoma. Front Immunol 2025; 16:1575627. [PMID: 40433373 PMCID: PMC12106304 DOI: 10.3389/fimmu.2025.1575627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Background The COMPASSION-15 trial demonstrated that cadonilimab plus chemotherapy (CAD-CHM) confers clinical benefits over placebo plus chemotherapy (PLA-CHM) as a first-line treatment for human epidermal growth factor receptor 2 (HER2)-negative advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, the introduction of cadonilimab substantially elevates treatment costs, and its cost-effectiveness relative to PLA-CHM remains undetermined. This study evaluates the cost-effectiveness of CAD-CHM compared with PLA-CHM from the perspective of the Chinese healthcare system. Methods A Markov model with three health states was developed to assess the cost-effectiveness of CAD-CHM in HER2-negative advanced G/GEJ adenocarcinoma. Clinical efficacy data were sourced from the COMPASSION-15 trial, while drug costs were calculated based on national tender prices, and additional costs and utility values were extracted from published literature. The analysis encompassed the overall population, as well as subgroups stratified by programmed death ligand 1 (PD-L1) combined positive score (CPS) ≥ 5 and CPS < 5. Outcomes included total costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios (ICERs). Sensitivity analyses were conducted to evaluate model robustness. Results The ICER of CAD-CHM was $67,378.09 per QALY in the overall population, $48,433.34 per QALY in the PD-L1 CPS ≥ 5 subgroup, and $78,463.86 per QALY in the PD-L1 CPS < 5 subgroup. Key determinants influencing model outcomes included patient weight, cadonilimab cost, and the utility value of progression-free survival. Across all groups, CAD-CHM resulted in an ICER exceeding the willingness-to-pay threshold of $41,511 per QALY, with a 0% probability of cost-effectiveness compared with PLA-CHM. Conclusion From the perspective of the Chinese healthcare system, CAD-CHM is not cost-effective as a first-line treatment for HER2-negative advanced G/GEJ adenocarcinoma, either in the overall population or in subgroups stratified by PD-L1 CPS status, compared with chemotherapy alone.
Collapse
Affiliation(s)
- Zhifeng Zhou
- Department of Pharmacy, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yanqing Yang
- Department of Clinical Nutrition, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shaofang Chen
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| | - Maojin You
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| |
Collapse
|
25
|
Eckert T, Walton C, Bell M, Small C, Rowland NC, Rivers C, Zukas A, Lindhorst S, Fecci P, Strickland BA. The Basis for Targeting the Tumor Macrophage Compartment in Glioblastoma Immunotherapy. Cancers (Basel) 2025; 17:1631. [PMID: 40427130 PMCID: PMC12110244 DOI: 10.3390/cancers17101631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Glioblastoma (GBM) remains the most aggressive primary brain tumor with limited treatment options. The immunosuppressive tumor microenvironment (TME), largely shaped by tumor-associated macrophages (TAMs), represents a significant barrier to effective immunotherapy. Objective: This review aims to explore the role of TAMs within the TME, highlighting the phenotypic plasticity, interactions with tumor cells, and potential therapeutic targets to enhance anti-tumor immunity. Findings: TAMs constitute a substantial portion of the TME, displaying functional plasticity between immunosuppressive and pro-inflammatory phenotypes. Strategies targeting TAMs include depletion, reprogramming, and inhibition of pro-tumor signaling pathways. Preclinical studies show that modifying TAM behavior can shift the TME towards a pro-inflammatory state, enhancing antitumor immune responses. Clinical trials investigating inhibitors of TAM recruitment, polarization, and downstream signaling pathways reveal promising yet limited results, necessitating further research to optimize approaches. Conclusions: Therapeutic strategics targeting TAM plasticity through selective depletion, phenotypic reprogramming, or modulation of downstream immunosuppressive signals represent promising avenues to overcome GBM-associated immunosuppression. Early clinical trials underscore their safety and feasibility, yet achieving meaningful clinical efficacy requires deeper mechanistic understanding and combinatorial approaches integrating macrophage-direct therapies with existing immunotherapeutic modalities.
Collapse
Affiliation(s)
- Thomas Eckert
- School of Medicine, University of South Carolina, Columbia, SC 29209, USA
- MUSC Institute for Neuroscience Discovery, Charleston, SC 29425, USA;
| | - Chase Walton
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (C.W.); (M.B.)
| | - Marcus Bell
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; (C.W.); (M.B.)
| | - Coulter Small
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (C.S.); (A.Z.); (S.L.); (B.A.S.)
| | - Nathan C. Rowland
- MUSC Institute for Neuroscience Discovery, Charleston, SC 29425, USA;
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (C.S.); (A.Z.); (S.L.); (B.A.S.)
| | - Charlotte Rivers
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alicia Zukas
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (C.S.); (A.Z.); (S.L.); (B.A.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Scott Lindhorst
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (C.S.); (A.Z.); (S.L.); (B.A.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Peter Fecci
- Department of Neurosurgery, Duke University, Durham, NC 27710, USA;
| | - Ben A. Strickland
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA; (C.S.); (A.Z.); (S.L.); (B.A.S.)
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
26
|
Quandt Z, Lucas A, Liang SI, Yang E, Stone S, Fadlullah MZH, Bayless NL, Marr SS, Thompson MA, Padron LJ, Bucktrout S, Butterfield LH, Tan AC, Herold KC, Bluestone JA, Anderson MS, Spencer CN, Young A, Connolly JE. Associations between immune checkpoint inhibitor response, immune-related adverse events, and steroid use in RADIOHEAD: a prospective pan-tumor cohort study. J Immunother Cancer 2025; 13:e011545. [PMID: 40355283 PMCID: PMC12083316 DOI: 10.1136/jitc-2025-011545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/17/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have led to enduring responses in subsets of patients with cancer. However, these responses carry the risk of immune-related adverse events (irAEs), which can diminish the overall benefit of ICI treatment. While associations between irAE development and overall survival have been increasingly documented, there is a need for further understanding of these connections in large prospective real-world cohorts. METHODS The Resistance Drivers for Immuno-Oncology Patients Interrogated by Harmonized Molecular Datasets (RADIOHEAD) study, a pan-tumor, prospective cohort of 1,070 individuals undergoing standard of care first-line ICI treatment, aims to identify factors driving irAEs and clinical response. Clinical data and longitudinal blood samples were collected prospectively at multiple time points from 49 community-based oncology clinics across the USA. Structured, harmonized clinical data underwent unbiased statistical analysis to uncover predictors of real-world overall survival (rwOS) and risk factors for irAEs. RESULTS Across 1,070 participants' treatment courses, RADIOHEAD accumulated over 4,500 clinical data points. Patients experiencing any irAE (25.4%, n=272) exhibited significantly improved rwOS in the pan-tumor cohort (n=1,028, HR=0.41, 95% CI=(0.31, 0.55)). This association persisted when adjusting for age and metastatic disease in multivariate time-dependent Cox proportional hazard analysis, and was consistent across major tumor subtypes, including lung cancer and melanoma. Skin and endocrine irAEs of any grade were strongly associated with improved rwOS (Cox proportional hazard analysis, skin, p=2.03e-05; endocrine, p=0.0006). In this real-world cohort, the irAE rate appeared lower than those reported in clinical trials. Patients receiving corticosteroids prior to initiation of ICI treatment had significantly worse survival outcomes than non-users (HR 1.37, p=0.0054), with a stronger association with systemic steroid use (HR 1.75, p=0.0022). The risk of irAE was increased by exposure to combination immunotherapy relative to monotherapy (OR 4.17, p=2.8e-7), zoster vaccine (OR 2.4, p=5.2e-05), and decreased by prior chemotherapy (OR 1.69, p=0.0005). CONCLUSION The RADIOHEAD cohort is a well-powered, real-world cohort that clearly demonstrates the association between irAE development with improved response and baseline steroid use with worse response to ICI treatment after adjustment for survival bias.
Collapse
Affiliation(s)
- Zoe Quandt
- Department of Medicine, Division of Endocrinology and Metabolism, UCSF, San Francisco, California, USA
- Diabetes Center, UCSF, San Francisco, California, USA
| | - Anastasia Lucas
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha I Liang
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - EnJun Yang
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha Stone
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Muhammad Zaki Hidayatullah Fadlullah
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Nicholas L Bayless
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Sara Siebel Marr
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | - Lacey J Padron
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Samantha Bucktrout
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Aik Choon Tan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Departments of Oncological Sciences and Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Yale University, New Haven, Connecticut, USA
| | | | - Mark S Anderson
- Department of Medicine, Division of Endocrinology and Metabolism, UCSF, San Francisco, California, USA
- Diabetes Center, UCSF, San Francisco, California, USA
| | | | - Arabella Young
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - John E Connolly
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
27
|
Li H, Xu Y, Jiao X, Xu Q, Peng Z, Tang Y, Zhang J, Huang B, Shen Y, Chang B, Xia B, Duan W, Wang D, Zhu L, An R, Zhang G, Tang Y, Huang J, Qiu H, Wang L, Huang Y, Li G, Qian J, Sun L, Zheng H, Lou G, Zhang Y, Chen Y, Lu L, Cheng Y, Liu J, Zhao W, Ji J, He A, Wang K, Yu G, Zhu H, Ma C, Yuan J, Wang X, Zhang H, Ma X, Cai C, Yin K, Xie H, Wang Y, Wang S, Li L, Zhou H, Wang J, Zhu J, Ma D, Gao Q. IBI310 plus sintilimab vs. placebo plus sintilimab in recurrent/metastatic cervical cancer: A double-blind, randomized controlled trial. MED 2025; 6:100573. [PMID: 39827881 DOI: 10.1016/j.medj.2024.100573] [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: 10/22/2024] [Revised: 11/27/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND It remains unclear whether adding CTLA-4 blockade to PD-1/PD-L1 blockade improves clinical outcomes in cervical cancer (CC). METHODS In this randomized, double-blind, placebo-controlled, phase 2 study (ClinicalTrials.gov: NCT04590599), patients with recurrent/metastatic CC (R/M CC) who experienced disease progression after or during platinum-based chemotherapy were enrolled from 37 centers across China and randomly assigned (1:1), stratified by PD-L1 expression and prior treatment lines, to receive either IBI310 plus sintilimab or placebo plus sintilimab intravenously every 3 weeks for 12 weeks, followed by sintilimab alone. The primary endpoint was the objective response rate (ORR). Pivotal secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety. FINDINGS 205 patients were randomized to receive IBI310-sintilimab (n = 103) or placebo-sintilimab (n = 102). The ORR difference between the IBI310-sintilimab arm (32.3%, 95% confidence interval [CI]: 23.3%-42.5%) and the placebo-sintilimab arm (23.5%, 95% CI: 15.5%-33.1%) was not significant (p = 0.17). IBI310-sintilimab and placebo-sintilimab exhibited median PFS values of 3.6 (95% CI: 2.7-6.3) and 4.2 months (95% CI: 2.8-6.2), respectively (hazard ratio [HR] = 0.91, 95% CI: 0.65-1.27; p = 0.58). The median OSs were 13.9 months (95% CI: 11.5-25.6) in the IBI310-sintilimab arm and 17.2 months (95% CI: 13.7-25.9) in the placebo-sintilimab arm (HR = 1.12, 95% CI: 0.79-1.58; p = 0.54). Adding IBI310 to sintilimab increased the incidence of grade ≥3 treatment-related adverse events (55% versus 19%). CONCLUSIONS Compared to single-agent PD-1/PD-L1 blockade, dual blockade of CTLA-4 and PD-1/PD-L1 did not significantly improve clinical outcomes in R/M CC. FUNDING This work was funded by Innovent Biologics (Suzhou).
Collapse
Affiliation(s)
- Huayi Li
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yu Xu
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xiaofei Jiao
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Qin Xu
- Department of Gynecology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Jinan District, Fuzhou 350000, China
| | - Zikun Peng
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ying Tang
- Gynecological Oncology Center, Chongqing University Cancer Hospital, Chongqing 400000, China
| | - Jieqing Zhang
- Department of Gynecologic Oncology, Guangxi Medical University Cancer Hospital, Nanning 530000, China
| | - Bowen Huang
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yiyang Shen
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Baoping Chang
- Department of Medical Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang 471000, China
| | - Bairong Xia
- Department of Gynecology and Oncology, Anhui Provincial Cancer Hospital, Hefei 230000, China
| | - Wei Duan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100000, China
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110000, China
| | - Lijing Zhu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210000, China
| | - Ruifang An
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710000, China
| | - Guonan Zhang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliated to School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Yaling Tang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Jianli Huang
- Department of Oncology Radiotherapy, Zhangzhou Municipal Hospital of Fujian Province/Zhangzhou Affiliated Hospital of Fujian Medical Hospital, Zhangzhou 363000, China
| | - Hui Qiu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Li Wang
- Department of Gynecological Oncology, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450000, China
| | - Yi Huang
- Department of Gynecologic Oncology, Hubei Cancer Hospital, Wuhan 430000, China
| | - Guiling Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jianhua Qian
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Li Sun
- Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518000, China
| | - Hong Zheng
- Department of Gynecology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Cancer Hospital, Beijing 100000, China
| | - Ge Lou
- Department of Gynecologic Oncology, Harbin Medical University Cancer Hospital, Harbin 150000, China
| | - Youzhong Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Liqin Lu
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Yan Cheng
- Department of Gynecological Radiation Oncology, Zhengzhou University First Affiliated Hospital, Zhengzhou 450000, China
| | - Jihong Liu
- Department of Gynecologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510000, China
| | - Weidong Zhao
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Hefei 230000, China
| | - Jianghai Ji
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou 061000, China
| | - Aiqin He
- Department of Gynecological Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong 226000, China
| | - Ke Wang
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300000, China
| | - Guohua Yu
- Department of Medical Oncology, Weifang People's Hospital, Weifang 261000, China
| | - Hong Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Cailing Ma
- Department of Gynecology, Xinjiang Medical University Affiliated First Hospital, Urumqi 830000, China
| | - Jianlin Yuan
- Department of Gynecology and Obstetrics, Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi 830000, China
| | - Xia Wang
- Department of Oncology Radiotherapy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Hongfei Zhang
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xinyan Ma
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Chujun Cai
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Kang Yin
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Han Xie
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ya Wang
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Shuyan Wang
- Medical Science and Strategy Oncology, Innovent Biologics, Inc., Shanghai 200000, China
| | - Li Li
- Department of Biostatistics and Information, Innovent Biologics, Inc., Shanghai 200000, China
| | - Hui Zhou
- Medical Science and Strategy Oncology, Innovent Biologics, Inc., Shanghai 200000, China
| | - Jing Wang
- Department of Gynecologic Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| | - Jianqing Zhu
- Department of Gynecological Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310000, China.
| | - Ding Ma
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Qinglei Gao
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
28
|
Canali B, Apolone G, Ascierto PA, De Braud F, Grossi F, Perrone F, Fiorentino F, Di Costanzo A, Candelora L, Patanè G, Zapparelli G, Mezzanotte C, Didoni G, Riccaboni M. Effect of immuno-oncology on clinical and economic outcomes for a selection of cancers in Italy. Expert Rev Pharmacoecon Outcomes Res 2025:1-11. [PMID: 40329477 DOI: 10.1080/14737167.2025.2493130] [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: 10/09/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES This study assesses the impact of immune-oncology (IO) drugs' availability on cancer incidence-adjusted mortality rates from melanoma, lung, and renal cancers at population level in Italy between 2008 and 2019. METHODS We conducted a retrospective study on cross-sectional time-series aggregated data collected from publicly available sources and IQVIA proprietary databases. Three fixed-effects regression models were used to estimate how IO availability affects incidence-adjusted mortality for each cancer type. Estimated deaths were compared with deaths in a scenario with no IO drugs availability. Finally, the number of averted deaths was valued using the human capital approach. RESULTS A 1% increase in IO availability reduces incidence-adjusted mortality rates for melanoma, lung, and renal cancers by 0.125% (95% CI: 0.138-0.112; p < 0.01), 0.011% (95% CI: 0.013-0.009; p < 0.01) and 0.005% (95% CI: 0.006-0.003; p < 0.01) between the introduction of the drug in the therapeutic area and 2019. This reduction resulted in total savings of € 49.0 million, € 61.3 million, and € 10.9 million in indirect costs due to premature mortality, respectively. CONCLUSIONS IO drugs introduction in Italy between 2008 and 2019 was associated with a significant decrease in deaths from each cancer and, consequently, in savings in indirect costs related to premature mortality.
Collapse
Affiliation(s)
- Beatrice Canali
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Filippo De Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Grossi
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
- Medical Oncology Division, ASST Sette Laghi, Varese, Italy
| | - Francesco Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | | | | | - Laura Candelora
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | | | | | - Claudia Mezzanotte
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | - Guido Didoni
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | | |
Collapse
|
29
|
Kong SL, Feng Z, Kim S, Ha EK, Kamel K, Becich M, Luketich JD, Pennathur A. Hyperthermic Intrathoracic Chemoperfusion and the Role of Adjunct Immunotherapy for the Treatment of Pleural Mesothelioma. Biomolecules 2025; 15:678. [PMID: 40427571 PMCID: PMC12108701 DOI: 10.3390/biom15050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pleural mesothelioma (PM) is an aggressive cancer originating from the mesothelial lining of the pleura, with a rising global incidence since the mid-20th century due to asbestos and erionite exposure. PM accounts for 80-90% of all mesothelioma cases and is histologically classified into three subtypes-epithelioid, sarcomatoid, and biphasic- with epithelioid carrying the most favorable prognosis. Despite advances in surgery, chemotherapy, radiotherapy, and immunotherapy, PM prognosis remains poor, necessitating more effective, multimodal strategies. Hyperthermic intrathoracic chemoperfusion (HITHOC) has emerged as a promising adjunct to cytoreductive surgery by delivering heated chemotherapy directly to the pleural cavity, potentially improving survival-especially in patients with epithelioid PM. Combining HITHOC with post-surgical immunotherapy represents a novel approach to enhancing both local and systemic anti-tumor responses and targeting microscopic disease and distant metastases. This review explores surgical outcomes after surgery for PM, the therapeutic synergy of HITHOC and immunotherapy, ongoing clinical trials evaluating this multimodal strategy, and its implications for future patient care.
Collapse
Affiliation(s)
- Susan Luozheng Kong
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Zihan Feng
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Sangmin Kim
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Edra K. Ha
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Kero Kamel
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Michael Becich
- Department of Bioinformatics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
- UPMC Hillman Cancer Center Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
30
|
Cartwright D, Kidd AC, Ansel S, Ascierto ML, Spiliopoulou P. Oncogenic Signalling Pathways in Cancer Immunotherapy: Leader or Follower in This Delicate Dance? Int J Mol Sci 2025; 26:4393. [PMID: 40362630 PMCID: PMC12072740 DOI: 10.3390/ijms26094393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Immune checkpoint inhibitors have become a mainstay of treatment in many solid organ malignancies. Alongside this has been the rapid development in the identification and targeting of oncogenic drivers. The presence of alterations in oncogenic drivers not only predicts response to target therapy but can modulate the immune microenvironment and influence response to immunotherapy. Combining immune checkpoint inhibitors with targeted agents is an attractive therapeutic option but overlapping toxicity profiles may limit the clinical use of some combinations. In addition, there is growing evidence of shared resistance mechanisms that alter the response to immunotherapy when it is used after targeted therapy. Understanding this complex interaction between oncogenic drivers, targeted therapy and response to immune checkpoint inhibitors is vital for selecting the right treatment, at the right time for the right patient. In this review, we summarise the preclinical and clinical evidence of the influence of four common oncogenic alterations on immune checkpoint inhibitor response, combination therapies, and the presence of shared resistance mechanisms. We highlight the common resistance mechanisms and the need for more randomised trials investigating both combination and sequential therapy.
Collapse
Affiliation(s)
- Douglas Cartwright
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Andrew C. Kidd
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Sonam Ansel
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Maria Libera Ascierto
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
| | - Pavlina Spiliopoulou
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK; (D.C.); (A.C.K.); (S.A.); (M.L.A.)
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| |
Collapse
|
31
|
Kovács SA, Kovács T, Lánczky A, Paál Á, Hegedűs ZI, Sayour NV, Szabó L, Kovács A, Bianchini G, Ferdinandy P, Ocana A, Varga ZV, Fekete JT, Győrffy B. Unlocking the power of immune checkpoint inhibitors: Targeting YAP1 reduces anti-PD1 resistance in skin cutaneous melanoma. Br J Pharmacol 2025. [PMID: 40324810 DOI: 10.1111/bph.70052] [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: 07/02/2024] [Revised: 02/02/2025] [Accepted: 03/15/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND PURPOSE Immune checkpoint inhibitors, such as anti-PD1, revolutionized melanoma treatment. However, resistance and low response rates remain problems. Our goal was to pinpoint actionable biomarkers of resistance to anti-PD1 treatment and verify therapeutic effectiveness in vivo. EXPERIMENTAL APPROACH Using receiver operating characteristic (ROC) and survival analysis in a database of 1434 samples, we identified the strongest resistance-associated genes. Inhibitors were evaluated in C57BL/6J mice using wild-type B16-F10, and BRAF, -PTEN, -CDKN2A-mutant YUMM1.7 melanoma cell lines. We investigated the synergistic impact of anti-PD1 therapy and yes-associated protein 1 (YAP1) inhibition by non-photoactivated Verteporfin. Tumour volume was determined at fixed cutoff points, normalized to body weights. KEY RESULTS In the anti-PD1-treated melanoma cohort, YAP1 was the strongest druggable candidate overexpressed in non-responder patients (ROC AUC = 0.699, FC = 1.8, P=1.1E-8). The baseline YAP1 expression correlated with worse progression-free survival (HR = 2.51, P=1.2E-6, FDR = 1%), and overall survival (HR = 2.15, P = 1.2E-5, FDR = 1%). In YUMM1.7, combination of Verteporfin plus anti-PD1 reduced tumour size more than anti-PD1 monotherapy (P=0.008), or control (P=0.021). There was no difference between the cohorts in B16-F10 inoculated mice. We found increased expression of YAP1 in YUMM1.7 mice compared to B16-F10. The combination therapy induced a more-immune-inflamed phenotype characterized by increased expression of T cell and M1 macrophage markers. CONCLUSIONS AND IMPLICATIONS Verteporfin with anti-PD1 exhibited antitumor potential by promoting a pro-inflammatory tumour microenvironment in melanoma. We believe that YAP1 acts as a master regulator of anti-PD1 resistance.
Collapse
Affiliation(s)
- Szonja Anna Kovács
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Oncology Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | | | - Ágnes Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsombor I Hegedűs
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Lilla Szabó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Andrea Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Alberto Ocana
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, Madrid, Spain
- Experimental Therapeutics in Cancer Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - János Tibor Fekete
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Oncology Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Győrffy
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
32
|
Ji C, Kumpf S, Qian J, Federspiel JD, Sheehan M, Capunitan D, Atallah E, Astbury S, Arat S, Oziolor E, Ocana MF, Ramaiah SK, Grove J, Aithal GP, Lanz TA. Transcriptomic and proteomic characterization of cell and protein biomarkers of checkpoint inhibitor-induced liver injury. Cancer Immunol Immunother 2025; 74:190. [PMID: 40317333 PMCID: PMC12049347 DOI: 10.1007/s00262-025-04033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/24/2025] [Indexed: 05/07/2025]
Abstract
Immune checkpoint inhibitors (ICI) targeting CTLA-4 and PD-1 have shown remarkable antitumor efficacy, but can also cause immune-related adverse events, including checkpoint inhibitor-induced liver injury (ChILI). This multi-omic study aimed to investigate changes in blood samples from treated cancer patients who developed ChILI. PBMCs were sequenced for by transcriptomic and T cell receptor repertoire (bulk and single-cell immune profiling), and extracellular vesicle (EV) enrichment from plasma was analyzed by mass spectroscopy proteomics. Data were analyzed by comparing the ChILI patient group to the control group who did not develop ChILI and by comparing the onset of ChILI to pre-ICI treatment baseline. We identified significant changes in T cell clonality, gene expression, and proteins in peripheral blood mononuclear cells (PBMCs) and plasma in response to liver injury. Onset of ChILI was accompanied by an increase in T cell clonality. Pathway analysis highlighted the involvement of innate and cellular immune responses, mitosis, pyroptosis, and oxidative stress. Single-cell RNA sequencing revealed that these changes were primarily found in select T cell subtypes (including CD8 + effector memory cells), while CD16 + monocytes exhibited enrichment in metabolic pathways. Proteomic analysis of plasma extracellular vesicles showed enrichment in liver-associated proteins among differentially expressed proteins. Interestingly, an increase in PBMC PD-L1 gene expression and plasma PD-L1 protein was also found to be associated with ChILI onset. These findings provide valuable insights into the immune and molecular mechanisms underlying ChILI as well as potential biomarkers of ChILI.Trial registration number NCT04476563.
Collapse
Affiliation(s)
- Changhua Ji
- Drug Safety R&D, Pfizer Inc, 10777 Science Center Dr., La Jolla, CA, 92121, USA.
| | - Steven Kumpf
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | - Jessie Qian
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | | | - Mark Sheehan
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | - Darien Capunitan
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | - Edmond Atallah
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Stuart Astbury
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Seda Arat
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | - Elias Oziolor
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA
| | | | | | - Jane Grove
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Thomas A Lanz
- Drug Safety R&D, Pfizer Inc, Eastern Point Rd, 274-3715A, Groton, CT, 06340, USA.
| |
Collapse
|
33
|
Reinhard S, Utikal JS, Zaremba A, Lodde G, von Wasielewski I, Klespe KC, Meier F, Haferkamp S, Kähler KC, Herbst R, Gebhardt C, Sindrilaru A, Dippel E, Angela Y, Mohr P, Pfoehler C, Forschner A, Kaatz M, Schell B, Gesierich A, Loquai C, Hassel JC, Ulrich J, Meiss F, Schley G, Heinzerling LM, Sachse M, Welzel J, Weishaupt C, Sunderkötter C, Michl C, Lindhof HH, Kreuter A, Heppt MV, Wenk S, Mauch C, Berking C, Nedwed AS, Gutzmer R, Leiter U, Schadendorf D, Ugurel S, Weichenthal M, Haist M, Fleischer MI, Lang B, Grabbe S, Stege H. First-line checkpoint inhibitor therapy in metastatic acral lentiginous melanoma compared to other types of cutaneous melanoma: A multicenter study from the prospective skin cancer registry ADOREG. Eur J Cancer 2025; 220:115356. [PMID: 40121837 DOI: 10.1016/j.ejca.2025.115356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Melanoma is the main cause of skin cancer-related death. Treatment with immune checkpoint inhibitors (CPI) has improved the prognosis in recent years. However, subtypes of melanoma differ in their response. Acral lentiginous melanoma (ALM) has a worse prognosis compared to cutaneous melanoma other than ALM (CM) and is therefore of particular relevance. AIMS To evaluate the efficacy of CPI in first-line treatment of patients with advanced ALM compared CM. METHODS Retrospective analysis of patients with metastatic ALM (n = 45) or CM (n = 328) who received first-line CPI therapy from the multicenter prospective skin cancer registry ADOREG. Study endpoints were best overall response (BOR), progression-free survival (PFS) and overall survival (OS). RESULTS ALM patients had significantly higher rates of ulcerated tumors, loco regional metastases and fewer BRAF-mutated tumors compared to CM patients. Combined CPI was administered in 48.9 % ALM patients and 39.3 % of CM patients, while the remaining patients received PD-1 monotherapy. OS trended to be shorter in patients with ALM (18.1 vs. 43.8 months, p = 0.10) with no significant differences in PFS (7.0 vs. 11.5 months, p = 0.21). In patients with CM, median OS with combined CPI was not reached, whereas the median OS after PD-1 monotherapy was 37.8 months (p = 0.22). Conversely, in patients with ALM, OS with combined CPI was 17.8 months, compared to 26 months with PD-1 monotherapy (p = 0.15). There were no significant differences in BOR between patients with ALM or CM. CONCLUSION Analysis of this real-world cohort of patients with metastatic melanoma showed a trend towards poorer survival outcomes upon first-line treatment with CPI in ALM compared to cutaneous melanoma of other subtypes.
Collapse
Affiliation(s)
- Sören Reinhard
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Anne Zaremba
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Georg Lodde
- Department of Dermatology, Venereology and Allergology, University Hospital Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Imke von Wasielewski
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Kai Christian Klespe
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Katharina C Kähler
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Rudolf Herbst
- Department of Dermatology, HELIOS Hospital Erfurt, Erfurt, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anca Sindrilaru
- Department of Dermatology, University Hospital of Ulm, Ulm, Germany
| | - Edgar Dippel
- Department of Dermatology, Ludwigshafen City Hospital, Ludwigshafen, Germany
| | - Yenny Angela
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Claudia Pfoehler
- Department of Dermatology, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Martin Kaatz
- Department of Dermatology, DRK Hospital Chemnitz-Rabenstein, Rabenstein, Germany
| | - Beatrice Schell
- Department of Dermatology, Wald-Klinikum Gera, Gera 07546, Germany
| | - Anja Gesierich
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Carmen Loquai
- Department of Dermatology, Klinikum Bremen-Ost, Gesundheit Nord gGmbH, Bremen, Germany
| | - Jessica C Hassel
- Heidelberg University, Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jens Ulrich
- Department of Dermatology and Allergy, Harzklinikum Dorothea Christiane Erxleben GmbH, Quedlinburg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gaston Schley
- Department of Dermatology, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Lucie M Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilian Universität Munich, München, Germany
| | - Michael Sachse
- Department of Dermatology, Hospital Bremerhaven Reinkenheide, Bremerhaven, Germany
| | - Julia Welzel
- Department of Dermatology, University Hospital Augsburg, Augsburg, Germany
| | - Carsten Weishaupt
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Cord Sunderkötter
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Christiane Michl
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | - Alexander Kreuter
- Department of Dermatology, Venerology and Allergology, Helios St. Elisabeth Klinik Oberhausen, University Witten-Herdecke, Oberhausen, Germany
| | - Markus V Heppt
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Saskia Wenk
- Department of Dermatology, Medical Center Klinikum Darmstadt, Teaching Hospital Goethe-University Frankfurt, Darmstadt, Germany
| | - Cornelia Mauch
- Department of Dermatology, Ruhr-Universität Bochum, Bochum, Germany
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN) and CCC Alliance WERA, Bavarian Cancer Research Center (BZKF), Erlangen 91052, Germany
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Muehlenkreiskliniken Minden and Ruhr University Bochum, Minden, Germany
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard-Karls University of Tübingen, Tubingen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Michael Weichenthal
- Department of Dermatology, Skin Cancer Center, University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | | | - Berenice Lang
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Mainz 55131, Germany.
| |
Collapse
|
34
|
Gao M, Liu Y, Zhao L, Chen J, Wan W, Yuan Z, Li L, Huang Y, Wang Y, Zheng Y. Cell Surface-Tethered Nucleic Acid Therapeutics Program Robust and Tumor-Responsive Enhancement of Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419969. [PMID: 40318090 DOI: 10.1002/adma.202419969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/05/2025] [Indexed: 05/07/2025]
Abstract
The efficacy of adoptive T cell therapy (ACT) against solid tumors is significantly limited by the immunosuppressive tumor microenvironment (TME). Systemic administration of immunostimulants provides inadequate support to ACT cells and often elicits systemic toxicities. Here we present cell-surface-anchored nucleic acid therapeutics (NATs) to robustly enhance ACT through synergistic blockade of immunosuppressive adenosine and PD-1/PD-L1 pathways in tumors. Two distinct NATs-DNA aptamers targeting PD-L1 (aptPD-L1) and ATP (aptATP)-are engineered to form partially-hybridized duplexes (aptDual) that can efficiently anchor to cell surface before transfer. Backpacked aptDual spatial-temporally co-localize with ACT cells in vivo and jointly infiltrate the ATP-rich TME. Upon binding with ATP, aptDual dissociates to responsively release aptPD-L1. Concurrently, aptATP scavenges extracellular ATP and its metabolite adenosine to disrupt the inhibitory adenosinergic axis, thereby sensitizing ACT cells to immune checkpoint blockade by aptPD-L1. This dual inhibition elicited a remarkable 40-fold increase in functional tumor-infiltrating ACT cells, substantially boosting the efficacy of TCR-T and CAR-T cells in multiple solid tumor models, even in immunologically "cold" tumors. NAT backpacks provide a facile, versatile, and safe strategy to augment various ACTs against solid tumors.
Collapse
Affiliation(s)
- Mengqian Gao
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, 215123, P. R. China
| | - Yingyu Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310000, P. R. China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, P. R. China
| | - Lei Zhao
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, 215123, P. R. China
| | - Jin Chen
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
| | - Wenjun Wan
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, 215123, P. R. China
| | - Ze Yuan
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
| | - Lingyu Li
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
| | - Yulun Huang
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
| | - Yajun Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310000, P. R. China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, P. R. China
| | - Yiran Zheng
- The Fourth Affiliated Hospital of Soochow University, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215123, P. R. China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
35
|
Weichenthal M, Ellebaek E, Mangana J, Asher N, Gavrilova I, Kandolf L, Ugurel S, Hausschild A, Meier F, Leiter U, Livingstone E, Gebhardt C, Gutzmer R, Ruhlmann CH, Mahncke-Guldbrandt L, Haslund CA, Kopec S, Teterycz P, Bender M, Poudroux W, Muñoz-Couselo E, Berciano-Guerrero MA, Shalamanova G, DePalo DK, Brozić JM, Chiarion-Sileni V, Arance A, Ziogas D, Robert C, van de Velde AO, Gassama AA, Shapira R, BenBetzalel G, Grynberg S, Ramelyte E, Bertoldo F, DelPrete V, Gaudy-Marqueste C, Mohr P, Dummer R, Ascierto PA, Gogas H, Espinosa E, Lebbé C, Rutkowski P, Haanen J, Schadendorf D, Svane IM. Immune checkpoint inhibition in metastatic or non-resectable melanoma after failure of adjuvant anti-PD-1 treatment. A EUMelaReg real-world evidence study. Eur J Cancer 2025; 220:115339. [PMID: 40090216 DOI: 10.1016/j.ejca.2025.115339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Adjuvant immune checkpoint inhibition (ICI) with anti-PD-1 antibodies in high-risk resected melanoma has been shown to improve recurrence-free survival. It is unclear whether prior adjuvant anti-PD-1 therapy is associated with altered response to subsequent ICI treatment in the metastatic setting. METHODS Using data from the European Melanoma Registry (EUMelaReg), we analyzed the efficiency of first-line (1L) ICI in non-resectable or metastatic melanoma after failure from prior adjuvant anti-PD-1 treatment. Both single-agent anti-PD-1 and combined anti-PD-1/CTLA-4 (Ipi/Nivo) 1L regimes were included in the analysis. We identified 389 patients receiving 1L ICI with prior adjuvant anti-PD-1 treatment. The control population was selected from a pool of 3390 PD-1-naive cases by 1:1 matching for the type of 1L ICI and various prognostic factors. As outcome measure, overall remission rates (ORR) were calculated and progression-free survival (PFS) was evaluated by Kaplan-Meier and Cox regression analysis. RESULTS Out of 389 patients, 303 (77.9 %) received Ipi/Nivo and 86 (22.1 %) anti-PD-1 in 1L. ORR was significantly lower in pre-treated patients (31.4 %) as compared to anti-PD-1 naive patients (48.8 %; p < 0.0001). Kaplan-Meier analysis showed significantly shorter median PFS for pre-treated patients. This applied to both anti-PD-1 and Ipi/Nivo treatment. Patients with early recurrence from adjuvant treatment (during or up to 12 weeks after end of treatment) showed lower ORR (28.5 %) and shorter PFS (3.1 months) than those who recurred later (37.7 % and 6.1 months, respectively). CONCLUSIONS Patients with metastatic melanoma, previously exposed to anti-PD-1 ICI in the adjuvant setting showed significantly lower ORR and shorter PFS to 1L ICI with either Ipi/Nivo or single-agent anti-PD-1 retreatment.
Collapse
Affiliation(s)
- Michael Weichenthal
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Nethanel Asher
- Skin Cancer and Melanoma Center at Davidoff Cancer Center, Rabin Medical Center, Israel
| | - Iva Gavrilova
- Oncodermatology Department, National Oncology Center, Sofia, Bulgaria
| | - Lidija Kandolf
- Department of Dermatology, Faculty of Medicine, Military Medical Academy, Belgrade, Serbia
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Axel Hausschild
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Leiter
- Universitäts-Hautklinik Tübingen, Tübingen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Skin Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Minden, Germany
| | | | | | | | - Sylwia Kopec
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Marc Bender
- Skin Cancer Center, Division of Molecular Cell Biology, Elbe Kliniken Stade-Buxtehude, Buxtehude, Germany
| | | | - Eva Muñoz-Couselo
- Medical oncology department, Vall d'Hebron Hospital, Barcelona-Spain and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Miguel-Angel Berciano-Guerrero
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Danielle K DePalo
- Department of Cutaneous Oncology, Surgical Oncology Research Fellow, Moffitt Cancer Center, Tampa, FL, United States
| | - Jasmina Marić Brozić
- Department of Oncology, UHC Sestre milosrdnice, School of Medicine Zagreb, Zagreb, Croatia
| | | | - Anna Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Dimitrios Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Caroline Robert
- Gustave Roussy Cancer Campus and Université Paris Saclay, Villejuif, France
| | | | - Awa Aminata Gassama
- Scientific Administration, Biometrics Department, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ronnie Shapira
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Guy BenBetzalel
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Shirly Grynberg
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| | - Fabio Bertoldo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Valerio DelPrete
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Caroline Gaudy-Marqueste
- Aix-Marseille Univ, APHM, Hôpital Timone, Dermatology and Skin Cancer Department. Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseilles, France
| | - Peter Mohr
- Department of Dermatology, Elbekliniken Buxtehude, Buxtehude, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Paolo A Ascierto
- Department of Skin Cancers, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Enrique Espinosa
- Service of Oncology, Hospital U. La Paz, U. Autónoma de Madrid, Madrid-CIBERONC, Spain
| | - Celeste Lebbé
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer Institute APHP, INSERM U976, Saint Louis Hospital, Paris, France.
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - John Haanen
- Netherlands Cancer Institut Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
36
|
Wang SJ, Xiu J, Butcher KM, DeClerck BK, Kim GH, Moser J, Gibney GT, Hernandez‐Aya LF, Lutzky J, Abdulla F, Margolin KA, Possik PA, Robles‐Espinoza CD, Ito F, In GK. Comprehensive Profiling of Acral Lentiginous Melanoma Reveals Downregulated Immune Activation Compared to Cutaneous Melanoma. Pigment Cell Melanoma Res 2025; 38:e70027. [PMID: 40405404 PMCID: PMC12099029 DOI: 10.1111/pcmr.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2025] [Revised: 04/27/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025]
Abstract
Acral lentiginous melanoma (ALM) is a rare and insufficiently understood subtype of melanoma lacking in effective treatment options. Recent work has demonstrated that the response of ALM to immune checkpoint blockade is inferior to that of cutaneous melanoma (CM). Here we performed bulk genomic and transcriptomic sequencing of tumor tissue from 28 ALM and 5692 CM cases. Similar to prior studies, ALM was associated with a significantly lower incidence of point mutations, including in the TERT promoter and BRAF, but increased numbers of gene amplifications, notably of CCND1, HMGA2, and MDM2. Reactome pathway analysis revealed enhancement of keratinization and PI3K/AKT signaling pathways. Overall immunogenicity was decreased in ALM, which possessed lower IFNγ (p < 0.001) and T-cell inflammatory (p = 0.03) pathway scores than CM. Despite higher computationally inferred levels of myeloid dendritic cells (p = 0.006), neoantigen load independent of predicted HLA binding affinity was lower (p < 0.01) in ALM versus CM. Assessment of classical and nonclassical HLA mRNA levels revealed upregulation of HLA-G, suggesting alternative ALM immune evasion pathways in the setting of lower PD-L1 expression (p = 0.005). Additional research is needed to better understand and therapeutically target signaling networks in the ALM tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie J. Wang
- Department of MedicineUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | | | - Katherine M. Butcher
- Division of OncologyUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Brittney K. DeClerck
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Department of PathologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | - Gene H. Kim
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Department of PathologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| | - Justin Moser
- HonorHealth Research and Innovation InstituteScottsdaleArizonaUSA
| | | | | | - Jose Lutzky
- University of Miami Sylvester Comprehensive Cancer CenterMiamiFloridaUSA
| | | | - Kim A. Margolin
- Department of Medical OncologySaint John's Cancer Institute, Providence Saint John's Health CancerSanta MonicaCaliforniaUSA
| | - Patrícia Abrão Possik
- Division of Basic and Experimental ResearchBrazilian National Cancer InstituteRio de JaneiroBrazil
| | | | - Fumito Ito
- Department of SurgeryUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Gino K. In
- Department of MedicineUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
- Division of OncologyUniversity of Southern California Keck School of Medicine, Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
- Department of DermatologyUniversity of Southern California Keck School of MedicineLos AngelesCaliforniaUSA
| |
Collapse
|
37
|
Ong CV, Samlowski W. Timing of brain metastases in relation to outcome during first-line ipilimumab plus nivolumab therapy for metastatic melanoma in a community oncology practice. J Neurooncol 2025; 172:645-653. [PMID: 39932529 PMCID: PMC11968462 DOI: 10.1007/s11060-025-04951-z] [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: 11/25/2024] [Accepted: 01/23/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE Patients with metastatic melanoma frequently develop brain metastases. Due to recent advances in melanoma therapy, we evaluated the timing of brain metastases diagnosis in relation to outcome during melanoma immunotherapy. METHODS Patients who received 1st -line treatment with ipilimumab plus nivolumab for metastatic melanoma were identified via a database search. Patient characteristics and outcomes were recorded. RESULTS Of 73 patients that met study criteria, 20 patients developed brain metastases (27.4%). Of these 20 patients, 14 had brain metastases at diagnosis of metastatic disease, Only 6 progressed in the brain following immunotherapy. All but one patient with brain metastases at diagnosis were symptomatic. Following immunotherapy, 4/15 (all with BRAF V600E mutations) achieved complete remissions and prolonged survival. Each of these patients was able to undergo elective treatment discontinuation. One additional patient developed stable disease. Delayed brain metastases proved to be infrequent (6/59 patients). Delayed brain metastases were always diagnosed within the first 15 months of treatment. Five of these 6 patients died, with a median progression-free survival of only 2.1 months. CONCLUSION Brain metastases frequently complicated the course of metastatic melanoma. Patients with symptomatic brain metastases at diagnosis had a potential for durable remissions following multidisciplinary treatment, particularly if a BRAF V600E mutation was present. This included 2 of 10 patients who were on steroid treatment prior to the start of immunotherapy. Treatment with combination immunotherapy seemed to reduce the development of subsequent brain metastases. Patients who developed delayed brain metastases had a very poor outlook, despite attempted salvage therapy.
Collapse
Affiliation(s)
- Claire Victoria Ong
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV, USA
| | - Wolfram Samlowski
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, NV, USA.
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA.
- Department of Internal Medicine, University of Nevada School of Medicine, Reno, NV, USA.
- Comprehensive Cancer Centers of Nevada, 9280 W. Sunset Rd., Suite 100, Las Vegas, NV, 89148, USA.
| |
Collapse
|
38
|
Wang H, Zhou R, Xu C, Dai L, Hou R, Zheng L, Fu C, Shi G, Wang J, Li Y, Cen J, Xu X, Yu L, Li Y, Wang J, Du Q, Li Z. GRP78 Nanobody-Directed Immunotoxin Activates Innate Immunity Through STING Pathway to Synergize Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408086. [PMID: 40135833 PMCID: PMC12097070 DOI: 10.1002/advs.202408086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/27/2025] [Indexed: 03/27/2025]
Abstract
The lack of targetable antigens poses a significant challenge in developing effective cancer-targeted therapies. Cell surface translocation of endoplasmic reticulum (ER) chaperones, such as glucose-regulated protein 78 (GRP78), during malignancy, drug resistance, and ER stress induced by therapies, offers a promising pan-cancer target. To target GRP78, nanobody C5, identified from a phage library and exhibiting high affinity for human and mouse GRP78, is utilized to develop the Pseudomonas exotoxin (PE) immunotoxin C5-PE38. C5-PE38 induced ER stress, apoptosis and immunogenic cell death in targeted cells and showed antitumor efficacy against colorectal cancer and melanoma models without obvious toxicity. Mechanistically, transcriptome profiling showed that C5-PE38 reshaped the tumor immune microenvironment with enhanced innate and adaptive immune response and response to interferon beta. Moreover, C5-PE38-induced cell death could trans-activate STING pathway in dendritic cells and macrophages, promoting CD8+ T cell infiltration. It also sensitizes both primary and metastatic melanomas to anti-PD1 therapy, partly through STING activation. Overall, this study unveils a feasible GRP78 nanobody-directed therapy strategy for single or combinatorial cancer intervention. This work finds that C5-PE38-induced cell death stimulates STING-dependent cytosolic DNA release to promote antitumor immunity, a mechanism not previously reported for PE38, providing valuable insights for its clinical use.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Post‐doctoral Scientific Research Station of Basic MedicineJinan UniversityGuangzhou510632China
| | - Runhua Zhou
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chengchao Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Lingyun Dai
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Rui Hou
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Harry Perkins Institute of Medical ResearchQEII Medical Centre and Centre for Medical ResearchThe University of Western AustraliaNedlandsWA6009Australia
| | - Liuhai Zheng
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Chunjin Fu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Guangwei Shi
- Department of Neurosurgery & Medical Research CenterShunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)Guangzhou510515China
| | - Jingwei Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yang Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Jinpeng Cen
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaolong Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Le Yu
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yilei Li
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jigang Wang
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsArtemisinin Research CenterInstitute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifeng475004China
| | - Qingfeng Du
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhijie Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| |
Collapse
|
39
|
Siefker-Radtke AO, Huddart RA, Bilen MA, Balar A, Castellano D, Sridhar SS, De Giorgi U, Penkov K, Vasiliev A, Peer A, Järvinen R, Harputluoğlu H, Koshkin VS, Poushnejad S, Wang T, Qureshi A, Tagliaferri MA, Zalevsky J, Loriot Y. Bempegaldesleukin plus nivolumab in first-line advanced/metastatic urothelial carcinoma: Results from a phase II single-arm study (PIVOT-10). Urol Oncol 2025; 43:330.e1-330.e9. [PMID: 39477771 DOI: 10.1016/j.urolonc.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 05/14/2025]
Abstract
BACKGROUND In PIVOT-02, bempegaldesleukin (BEMPEG), a pegylated interleukin-2 cytokine prodrug, in combination with nivolumab (NIVO), a Programmed cell death protein 1 inhibitor, demonstrated the potential to provide additional benefits over immune checkpoint inhibitor monotherapy in patients with urothelial carcinoma, warranting further investigation. We evaluated BEMPEG plus NIVO in cisplatin-ineligible patients with previously untreated locally advanced or metastatic urothelial carcinoma. METHODS This open-label, multicenter, single-arm, phase II study enrolled patients with locally advanced/surgically unresectable or metastatic urothelial carcinoma and who were ineligible for cisplatin-based treatment. Patients received BEMPEG plus NIVO were administered intravenously every 3 weeks for ≤2 years or until progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR) by blinded independent central review (BICR) in patients with low programmed death ligand-1 (PD-L1) expression. Secondary endpoints included ORR and duration of response in the overall population. Progression-free survival (PFS) and overall survival (OS) were exploratory endpoints. RESULTS One hundred and eighty-eight patients were enrolled; 123 patients were PD-L1 low (combined positive score [CPS] <10; 65.4%), 59 were PD-L1 high (31.4%; CPS ≥10), and 6 had PD-L1 status unknown (3.2%). ORR per blinded independent central review in patients with PD-L1-low tumors was 17.9% (95% confidence interval [CI] 11.6-25.8) while in all treated patients was 19.7% (95% CI 14.3-26.1). Median PFS and OS in the overall population were 3.0 months and 12.6 months, respectively. BEMPEG plus NIVO combination was well tolerated, with a safety profile similar to previously reported trials; no new or unexpected safety signals were reported. CONCLUSIONS BEMPEG plus NIVO did not meet the efficacy threshold for ORR in patients with previously untreated locally advanced or metastatic urothelial carcinoma and low PD-L1 expression.
Collapse
Affiliation(s)
| | | | - Mehmet A Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA
| | - Arjun Balar
- Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | | | | | - Ugo De Giorgi
- Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Emilia-Romagna, Italy
| | - Konstantin Penkov
- Private Medical Institution Euromedservice, St. Petersburg, Russian Federation
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [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] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
41
|
Matsuura H, Ishino T, Ninomiya T, Ninomiya K, Tachibana K, Honobe‐Tabuchi A, Muto Y, Inozume T, Ueda Y, Ohashi K, Maeda Y, Nagasaki J, Togashi Y. High Antigenicity for T reg Cells Confers Resistance to PD-1 Blockade Therapy via High PD-1 Expression in T reg Cells. Cancer Sci 2025; 116:1214-1226. [PMID: 40017103 PMCID: PMC12044662 DOI: 10.1111/cas.70029] [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: 10/22/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Regulatory T (Treg) cells have an immunosuppressive function, and programmed death-1 (PD-1)-expressing Treg cells reportedly induce resistance to PD-1 blockade therapies through their reactivation. However, the effects of antigenicity on PD-1 expression in Treg cells and the resistance to PD-1 blockade therapy remain unclear. Here, we show that Treg cells gain high PD-1 expression through an antigen with high antigenicity. Additionally, tumors with high antigenicity for Treg cells were resistant to PD-1 blockade in vivo due to PD-1+ Treg-cell infiltration. Because such PD-1+ Treg cells have high cytotoxic T lymphocyte antigen (CTLA)-4 expression, resistance could be overcome by combination with an anti-CTLA-4 monoclonal antibody (mAb). Patients who responded to combination therapy with anti-PD-1 and anti-CTLA-4 mAbs sequentially after primary resistance to PD-1 blockade monotherapy showed high Treg cell infiltration. We propose that the high antigenicity of Treg cells confers resistance to PD-1 blockade therapy via high PD-1 expression in Treg cells, which can be overcome by combination therapy with an anti-CTLA-4 mAb.
Collapse
Affiliation(s)
- Hiroaki Matsuura
- Department of Tumor MicroenvironmentOkayama UniversityOkayamaJapan
- Department of Hematology, Oncology and Respiratory MedicineOkayama UniversityOkayamaJapan
| | - Takamasa Ishino
- Department of Tumor MicroenvironmentOkayama UniversityOkayamaJapan
| | | | - Kiichiro Ninomiya
- Department of Hematology, Oncology and Respiratory MedicineOkayama UniversityOkayamaJapan
| | - Kota Tachibana
- Department of Dermatology, Faculty of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | | | - Yoshinori Muto
- Department of DermatologyUniversity of YamanashiYamanashiJapan
| | - Takashi Inozume
- Department of DermatologyUniversity of YamanashiYamanashiJapan
- Department of DermatologyChiba UniversityChibaJapan
| | - Youki Ueda
- Department of Tumor MicroenvironmentOkayama UniversityOkayamaJapan
| | - Kadoaki Ohashi
- Department of Hematology, Oncology and Respiratory MedicineOkayama UniversityOkayamaJapan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory MedicineOkayama UniversityOkayamaJapan
| | - Joji Nagasaki
- Department of Tumor MicroenvironmentOkayama UniversityOkayamaJapan
| | - Yosuke Togashi
- Department of Tumor MicroenvironmentOkayama UniversityOkayamaJapan
- Department of Allergy and Respiratory MedicineOkayama University HospitalOkayamaJapan
- Kindai University Faculty of MedicineOsakaJapan
| |
Collapse
|
42
|
Lodde G, Albrecht LJ, Schadendorf D. [Treatment of metastatic melanoma: update 2025]. Dtsch Med Wochenschr 2025; 150:562-569. [PMID: 40262755 DOI: 10.1055/a-2500-0927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Immune checkpoint inhibition and targeted therapy with BRAF/MEK inhibition for BRAF-mutated melanoma have significantly improved progression-free and overall survival in patients with metastatic melanoma. Current research focuses on novel treatment strategies for PD-1 resistance, neoadjuvant approaches, and cellular therapies. 10-year follow-up data of randomized clinical trials show that both combined CTLA-4 and PD-1 immune checkpoint inhibition and PD-1 immune checkpoint inhibition alone can achieve long-term survival in metastatic melanoma. Potential surrogate markers of long-term response include a progression-free survival at 3 years after start of treatment and a reduction in tumour burden of at least 80%. The management of PD-1 resistance remains a challenge. Advances in molecular pathology have led to the identification of new therapeutic targets. Several cellular therapies are currently being evaluated in clinical trials as alternatives for melanoma patients refractory to immune checkpoint inhibition or targeted BRAF/MEK inhibition. In BRAF-mutant melanoma, combined BRAF/MEK inhibition is an alternative to immune checkpoint inhibition. Real-world data and clinical trial results on treatment sequencing suggest that immune checkpoint inhibition may improve survival in the first line setting, particularly in the absence of prior adjuvant systemic therapy. Adjuvant treatment leads to improved progression-free survival in melanoma patients while overall survival data are still pending. Neoadjuvant treatment seems to be a promising alternative to conventional adjuvant therapy for specific subgroups of melanoma patients. Participation in clinical trials offers patients the best opportunity to benefit from the latest treatment options.
Collapse
|
43
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
44
|
Xu Q, Chu J, Hu Q, Sun Y, Jiang F, Li S, Liu L. The role and clinical significance of tumor-draining lymph nodes in tumor progression and immunotherapy. Crit Rev Oncol Hematol 2025; 212:104745. [PMID: 40315968 DOI: 10.1016/j.critrevonc.2025.104745] [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: 03/18/2025] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) play a pivotal role in tumor growth and the immune response, activating immune cells such as CD8 + T cells and natural killer cells to combat tumors. However, tumors can subvert TDLNs to avoid immune attack. Initially, TDLNs stimulate a robust antitumor response, but as tumor evolve, they infiltrate with immunosuppressive cells that alter the TDLN environment and potentially promote metastasis. Immunotherapy, including immune checkpoint inhibitor (ICI), have emerged as a potential solution to this challenge by reconfiguring the TDLN environment to enhance immune responses and influence the immune status of the primary tumor. The integrity of the TDLNs is crucial for the efficacy of immunotherapy. Conventional surgery often removes TDLNs, but this may impede immune system function and the effectiveness of immunotherapy. It is therefore recommended that removal of TDLNs be considered after neoadjuvant treatment rather than before adjuvant treatment. Accurate identification of patients who require post-neoadjuvant TDLN removal and the determination of metastatic nodes is of paramount importance in tailoring treatment plans, optimizing of patient outcomes, and improving quality of life.
Collapse
Affiliation(s)
- Qian Xu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiahui Chu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qinqin Hu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanheng Sun
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fan Jiang
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Song Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lian Liu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
45
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [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: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
46
|
Szabó IL, Emri G, Ladányi A, Tímár J. Clinical Applications of the Molecular Landscape of Melanoma: Integration of Research into Diagnostic and Therapeutic Strategies. Cancers (Basel) 2025; 17:1422. [PMID: 40361349 PMCID: PMC12071057 DOI: 10.3390/cancers17091422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The molecular landscape of cutaneous melanoma is complex and heterogeneous, and a deeper understanding of the genesis and progression of the tumor driven by genetic alterations is essential for the development of effective diagnostic and therapeutic strategies. Molecular diagnostics and the use of biomarkers are increasingly playing a role in treatment decisions. However, further research is urgently needed to elucidate the relationships between complex genetic alterations and the effectiveness of target therapies (although BRAF mutation is still the only targeted genetic alteration). Further research is required to exploit other targetable genetic alterations such as NRAS, KIT or rare mutations. Treatment guidelines for cutaneous melanoma are continually evolving based on data from recent and ongoing clinical trials. These advancements reflect changes mainly in the optimal timing of systemic therapy and the choice of combination therapies increasingly tailored to molecular profiles of individual tumors. Mono- or combination immunotherapies demonstrated unprecedented success of melanoma treatment; still, there is room for improvement: though several factors of primary or acquired resistance are known, they are not part of patient management as biomarkers. The novel developments of cancer vaccines to treat melanoma (melanoma-marker-based or personalized neoantigen-based) are encouraging; introduction of them into clinical practice without proper biomarkers would be the same mistake made in the case of first-generation immunotherapies.
Collapse
Affiliation(s)
- Imre Lőrinc Szabó
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (I.L.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; (I.L.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Ladányi
- Department of Surgical and Molecular Pathology, National Institute of Oncology, 1122 Budapest, Hungary;
- National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary
| |
Collapse
|
47
|
Alsaafeen BH, Ali BR, Elkord E. Combinational therapeutic strategies to overcome resistance to immune checkpoint inhibitors. Front Immunol 2025; 16:1546717. [PMID: 40342408 PMCID: PMC12058545 DOI: 10.3389/fimmu.2025.1546717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Over the past few years, immune checkpoint inhibitors resulted in magnificent and durable successes in treating cancer; however, only a minority of patients respond favorably to the treatment due to a broad-spectrum of tumor-intrinsic and tumor-extrinsic factors. With the recent insights gained into the mechanisms of resistance, combination treatment strategies to overcome the resistance and enhance the therapeutic potential of immune checkpoint inhibitors are emerging and showing promising results in both pre-clinical and clinical settings. This has been derived through multiple interconnected mechanisms such as enhancing tumor immunogenicity, improving neoantigen processing and presentation in addition to augmenting T cell infiltration and cytotoxic potentials. In the clinical settings, several avenues of combination treatments involving immune checkpoint inhibitors were associated with considerable improvement in the therapeutic outcome in terms of patient's survival and tumor growth control. This, in turn, increased the spectrum of cancer patients benefiting from the unprecedented and durable effects of immune checkpoint inhibitors leading to their adoption as a first-line treatment for certain cancers. Moreover, the significance of precision medicine in cancer immunotherapy and the unmet demand to develop more personalized predictive biomarkers and treatment strategies are also highlighted in this review.
Collapse
Affiliation(s)
- Besan H. Alsaafeen
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics & Suzhou Municipal Key Lab of Biomedical Sciences and Translational Immunology, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
48
|
Wen J, Wang Y, Wang S, Liang Y, Hu X, Ou Q, Bao H, Zhao K, Wang Y. Genetic and transcriptional insights into immune checkpoint blockade response and survival: lessons from melanoma and beyond. J Transl Med 2025; 23:467. [PMID: 40269853 PMCID: PMC12020166 DOI: 10.1186/s12967-025-06467-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Integration of immune checkpoint inhibitors (ICIs) with non-immune therapies relies on identifying combinatorial biomarkers, which are essential for patient stratification and personalized treatment. METHODS We analyzed genomic and transcriptomic data from pretreatment tumor samples of 342 melanoma patients treated with ICIs to identify mutations and expression signatures associated with ICI response and survival. External validation and mechanistic exploratory analyses were conducted in two additional datasets to assess generalizability. RESULTS Responders were more likely to have received anti-PD-1 therapy rather than anti-CTLA-4 and exhibited a higher tumor mutation burden (both P < 0.001). Mutations in the dynein axonemal heavy chain (DNAH) family genes, specifically DNAH2 (P = 0.03), DNAH6 (P < 0.001), and DNAH9 (P < 0.01), were enriched in responders. The combined mutational status of DNAH 2/6/9 effectively stratified patients by progression-free survival (hazard ratio [HR]: 0.69; 95% confidence interval [CI] 0.51-0.92; P = 0.013) and overall survival (HR: 0.58; 95% CI 0.43-0.78; P < 0.001), with consistent association observed in the validation cohort (HR: 0.28; 95% CI 0.12-0.61; P < 0.001). DNAH-altered melanomas exhibited upregulation of chemokine signaling, cytokine-cytokine receptor interaction, and cell cycle-related pathways, along with elevated expression of immune-related signatures in interferon signaling, cytolytic activity, T cell function, and immune checkpoints. Using LASSO logistic regression, we identified a 26-gene composite signature predictive of clinical response, achieving an area under the curve (AUC) of 0.880 (95% CI 0.825-0.936) in the training dataset and 0.725 (95% CI 0.595-0.856) in the testing dataset. High-risk patients, stratified by the expression levels of a 13-gene signature, demonstrated significantly shorter overall survival in both datasets (HR: 3.35; P < 0.001; HR: 2.93; P = 0.002). CONCLUSIONS This analysis identified potential molecular determinants of response and survival to ICI treatment. Insights from melanoma biomarker research hold significant promise for translation into other malignancies, guiding individualized anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Jiaxin Wen
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanfeng Wang
- Department of Pathology, Beidahuang Industry Group General Hospital, Harbin, 150088, China
| | - Song Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, 210032, China
| | - Yuxin Liang
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xiaozhen Hu
- Department of Scientific Affairs, Mabwell (Shanghai) Biotech Co., Ltd., Beijing, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, 210032, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, 210032, China
| | - Kuo Zhao
- Day Care Ward, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300000, China.
| | - Youyu Wang
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
49
|
Jeon Y, Park JW, Lee SJ, Seol A, Kim Y, Lim JM, Choi SG, Gwak J, Lee E, Woo SM, Kim YH, Hwang DY, Seo S. Metal-Phenolic Coordination mediated Nanoemulsions for All-in-One Drug Delivery. ACS APPLIED BIO MATERIALS 2025; 8:3218-3226. [PMID: 40085534 DOI: 10.1021/acsabm.5c00037] [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: 03/16/2025]
Abstract
Combination chemotherapy is a promising strategy for cancer treatment, enhancing antitumor efficacy while minimizing drug resistance and mitigating the risk of single-drug overdose toxicity. Polymeric drug delivery carriers for combination chemotherapy have been developed; however, the synthetic process of amphiphilic polymers is time-consuming and laborious. The polymer entanglement-based drug encapsulation has been limited in achieving a high multidrug encapsulation efficiency because of the intrinsic preference for encapsulation of drugs upon their polarity. Herein, inspired by dynamic bonding and supramolecular assembly of metal-phenolic coordinate bonds at the oil/water interface, nanoemulsions were fabricated via a dropwise emulsion process. The emulsion interface was formulated by the coordinate bonds and created a colloidally stable emulsion with 50-100 nm in diameter for 3 weeks. These nanoemulsions enabled the coencapsulation of anticancer drugs, hydrophilic gemcitabine, and hydrophobic paclitaxel. Moreover, the treatment of dual-drug-encapsulated nanoemulsions reduced cellular viability (57.0 ± 0.0%) compared to that of gemcitabine only encapsulated (84.0 ± 9.9%) and paclitaxel only encapsulated (83.4 ± 7.2%) nanoemulsion treatment, demonstrating the potential of multidrug delivery carriers for synergistic combination therapy.
Collapse
Affiliation(s)
- Yeji Jeon
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Jun Woo Park
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Su Jin Lee
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Ayun Seol
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Yeojin Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Jeong Min Lim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Seong Gyu Choi
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Juyong Gwak
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eunji Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sang Myung Woo
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
- Department of Cancer Biomedical Science, The National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| | - Yun-Hee Kim
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
- Department of Cancer Biomedical Science, The National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Sungbaek Seo
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
50
|
Zhang N, Li J, Ren Y, Xu Y. Comprehensive pan-cancer analysis of PPP1R3G reveals its diagnostic, prognostic, and immunotherapeutic implications. Discov Oncol 2025; 16:530. [PMID: 40232629 PMCID: PMC12000506 DOI: 10.1007/s12672-025-02361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PPP1R3G, a regulatory subunit of protein phosphatase 1, plays a critical role in glycogen metabolism and has been implicated in various cancers. This study provides a comprehensive pan-cancer analysis of PPP1R3G, evaluating its expression, diagnostic and prognostic significance, and potential as a therapeutic target. METHODS We performed an extensive pan-cancer analysis of PPP1R3G using several databases to assess its expression and investigate its correlations with clinical outcomes. Our investigation included assessing PPP1R3G's impact on survival, its correlation with immune checkpoints and tumor stemness scores, and its prognostic significance. We also explored its relationship with immunomodulators, genomic profiles, and immunological characteristics, as well as its response to immunotherapy and involvement in various biological pathways. RESULTS PPP1R3G expression varied significantly across different cancers and correlated with both diagnostic and prognostic outcomes. Moreover, PPP1R3G was significantly linked to immune checkpoints, immunomodulators, prognosis, immunoregulatory genes, tumor stemness, cellular function, and immune infiltration across numerous cancer types. Further analysis of PPP1R3G-related gene enrichment, mutation profiles, RNA modifications, and genomic heterogeneity revealed that missense mutations were the predominant alteration affecting PPP1R3G. CONCLUSIONS Overall, the expression of PPP1R3G is closely associated with various cancers and may serve as a potential biomarker for cancer detection.
Collapse
Affiliation(s)
- Nie Zhang
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Jiaoyu Li
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Yanzhi Ren
- Department of Cardiology, Shizhong District People's Hospital, Zaozhuang, China
| | - Yahui Xu
- Graduate School of Anhui Medical University, Hefei, China.
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|