51
|
van Dijk B, Janssen JC, van Daele PLA, de Jonge MJA, Joosse A, Verheul HMW, Epker JL, van der Veldt AAM. From ICI to ICU: A systematic review of patients with solid tumors who are treated with immune checkpoint inhibitors (ICI) and admitted to the intensive care unit (ICU). Cancer Treat Rev 2025; 136:102936. [PMID: 40222269 DOI: 10.1016/j.ctrv.2025.102936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have improved the survival of patients with different solid tumors and even resulted in cure of metastatic disease. Since the introduction of ICIs, an increasing number of patients is admitted to the ICU for severe and potentially life-threatening immune related adverse events (irAEs). The outcome of patients who are admitted to the ICU because of severe irAEs is still unknown. The aim of this systematic review is to collect evidence on the outcomes of patients with solid tumors who are admitted to the ICU because of irAEs. METHODS Medline, Embase, Cochrane central register of controlled trials and Google Scholar were searched systematically from 1975 to 24 September 2024. Articles were only included when describing patients with solid tumors who were admitted to the ICU because of irAEs after treatment with ICIs. Two independent reviewers extracted the data and assessed the risk of bias. RESULTS A total of 183 articles were included: two prospective ICU population-based studies, four retrospective ICU population-based studies, 25 retrospective studies describing irAEs with incidental ICU admissions, one review of case reports, and 153 articles with a total of 177 case reports. The six ICU population-based studies contained a total of 169 patients who were admitted to the ICU due to irAEs. In these six studies, the most frequently reported irAEs were pneumonitis and neurological irAEs. Of these 169 patients, 26% of the patients died on the ICU and an additional 8% of patients in the three to six months thereafter due to irAEs or disease progression. In all 183 included articles, various irAEs were described and the reported mortality rate varied from 0 to 53%. CONCLUSION The potential favorable outcomes of both the solid tumors and irAEs will probably result in more need for ICU admissions. Prospective clinical trials are needed to optimize the treatment strategy of severe irAEs at the ICU. Based on the favourable outcomes after life-threatening irAEs, ICU admission should definitely be considered for patients with solid tumors who have life-threatening irAEs.
Collapse
Affiliation(s)
- Brigit van Dijk
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Joséphine C Janssen
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Oncological Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Paul L A van Daele
- Department of Clinical Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Jelle L Epker
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
52
|
Klümper N, Cox A, Sjödahl G, Roghmann F, Bolenz C, Hartmann A, Grünwald V, Faltas BM, Hölzel M, Eckstein M. Pre-treatment metastatic biopsy: a step towards precision oncology for urothelial cancer. Nat Rev Urol 2025; 22:256-267. [PMID: 39472646 DOI: 10.1038/s41585-024-00951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 05/10/2025]
Abstract
Early metastatic spread and clonal expansion of individual mutations result in a heterogeneous tumour landscape in metastatic urothelial cancer (mUC). Substantial molecular heterogeneity of common drug targets, such as membranous NECTIN4, FGFR3 mutations, PDL1 or immune phenotypes, has been documented between primary and metastatic tumours. However, translational and clinical studies frequently do not account for such heterogeneity and often investigate primary tumour samples that might not be representative in patients with mUC. We propose this as a potential factor for why many biomarkers for mUC have failed to be integrated into clinical practice. Fresh pre-treatment metastatic biopsies enable the capturing of prevailing tumour biology in real time. The characterization of metastatic tumour samples can improve response prediction to immunotherapy, the anti-NECTIN4 antibody-drug conjugate enfortumab vedotin and the FGFR inhibitor erdafitinib. Routine metastatic biopsy can thus improve the precision of identifying driver druggable alterations, thus improving treatment selection for patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany.
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Alexander Cox
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | - Gottfrid Sjödahl
- Department of Translational Medicine, Division of Urological Research, Lund University, Lund, Sweden
| | - Florian Roghmann
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- Department of Urology and Paediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumour Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Bishoy M Faltas
- Department of Hematology/Oncology, Weill-Cornell Medicine, New York, NY, USA
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
53
|
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
|
54
|
Hata T, Yamada T, Goto Y, Amano A, Negi Y, Watanabe S, Furuya N, Oba T, Ikoma T, Nakao A, Tanimura K, Taniguchi H, Yoshimura A, Fukui T, Murata D, Kaira K, Shiotsu S, Hibino M, Okada A, Chihara Y, Kawachi H, Kijima T, Takayama K. Regimen Selection for Chemoimmunotherapy in Nonsquamous Non-Small Cell Lung Cancer with Low PD-L1 Expression: A Multicenter Retrospective Cohort Study. Clin Lung Cancer 2025; 26:e190-e198.e4. [PMID: 39864962 DOI: 10.1016/j.cllc.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Although chemoimmunotherapy is recommended for advanced nonsquamous non-small cell lung cancer (NSCLC) with low programmed cell death ligand 1 (PD-L1) expression, no head-to-head comparisons of immune checkpoint inhibitors (ICIs) have been performed. Therefore, we compared the effect and safety of regimens in these patients to guide evidence-based treatment. METHODS This retrospective study included patients with advanced nonsquamous NSCLC with a PD-L1 tumor proportion score of 1% to 49% administered ICI combination platinum-based chemotherapy between May 2018 and May 2023 at 19 institutions in Japan. The main analysis compared survival outcomes and the incidence of grade ≥3 adverse events among regimens. RESULTS Among 316 included patients (median [range] age, 69 [36-89] years; 242 males; 41 never smokers), 200 (63%), 68 (22%), and 48 (15%) received chemotherapy combined with anti-programmed cell death protein 1 (PD-1), anti-PD-L1, and anti-PD-1/cytotoxic T-lymphocyte associated protein 4 (CTLA-4) antibodies, respectively. The median overall survival times were 28.6, 23.1, and 24.4 months (P = .41), and the median progression-free survival times were 9.4, 7.2, and 8.7 months (P = .28) in the anti-PD-1/Chemo, anti-PD-L1/Chemo and anti-PD-1/CTLA-4/Chemo groups, respectively. The anti-PD-1/CTLA-4/Chemo group had the lowest incidence of hematologic toxicity (P = .13) and the highest incidence of nonhematologic toxicity (P = .07). The incidence of grade ≥3 pneumonitis was significantly lower in the anti-PD-L1/Chemo group (P = .049). CONCLUSIONS Despite comparable survival benefits, adverse events differed among three regimens in patients with low PD-L1 expression. Notably, anti-PD-L1 antibody combination chemotherapy may reduce the risk of severe pneumonitis.
Collapse
Affiliation(s)
- Tae Hata
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yasuhiro Goto
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Akihiko Amano
- Department of Respiratory Medicine, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshiki Negi
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomohiro Oba
- Department of Respiratory Medicine, Saitama Red Cross Hospital, Saitama, Japan
| | - Tatsuki Ikoma
- Department of Thoracic Oncology, Kansai Medical University, Hirakata, Japan
| | - Akira Nakao
- Department of Respiratory Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Keiko Tanimura
- Department of Medical Oncology, Fukuchiyama City Hospital, Fukuchiyama, Japan
| | - Hirokazu Taniguchi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiro Yoshimura
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Tomoya Fukui
- Department of Respiratory Medicine, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Daiki Murata
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Shinsuke Shiotsu
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Makoto Hibino
- Department of Respiratory Medicine, Shonan Fujisawa Tokushukai Hospital, Fujisawa, Japan
| | - Asuka Okada
- Department of Respiratory Medicine, Saiseikai Suita Hospital, Suita, Japan
| | - Yusuke Chihara
- Department of Respiratory Medicine, Uji-Tokushukai Medical Center, Uji, Japan
| | - Hayato Kawachi
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
55
|
Wang C, Niu X, Xia T, Wang P, Wang Y, Zhang Z, Zhang J, Ju S, Xiao Z. Predicting c-KIT Inhibitor Efficacy in Patient-Derived Models of Sinonasal Mucosal Melanomas through Integrated Histogram Analysis of Whole-Tumor DKI, IVIM, and DCE-MRI. Clin Cancer Res 2025; 31:1686-1699. [PMID: 39937224 DOI: 10.1158/1078-0432.ccr-24-3765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE To evaluate whole-tumor histogram analysis of diffusion kurtosis imaging (DKI), intravoxel incoherent motion (IVIM), and dynamic contrast-enhanced MRI (DCE-MRI) in predicting the efficacy of imatinib, a c-KIT inhibitor, for treating patient-derived models derived from sinonasal mucosal melanomas (MM). EXPERIMENTAL DESIGN This study included 38 patients with histologically confirmed sinonasal MM, who underwent DKI, IVIM, and DCE-MRI. Patient-derived tumor xenograft models and precision-cut tumor slices were established to evaluate tumor response to imatinib. Whole-tumor histogram analysis was conducted on imaging parameters, and logistic regression models were applied to determine the predictive value of these metrics in differentiating responders from nonresponders. RESULTS Among the 38 patients with sinonasal MM, 12 were classified as responders and 26 as nonresponders based on patient-derived tumor xenograft and precision-cut tumor slice model responses to imatinib. The DKI model revealed significant differences in mean, median, 10th percentile, and 90th percentile values of Dk and K between responders and nonresponders (P < 0.05). The IVIM model indicated significant differences in 10th percentile and mean values of D, with kurtosis f being a strong predictor. The DCE-MRI model, using the 90th percentile Ktrans metric, demonstrated robust predictive performance, achieving an AUC of 0.89, with 80.77% specificity and 91.67% sensitivity. The combined logistic model integrating DKI, IVIM, and DCE-MRI metrics produced the highest predictive accuracy, with an AUC of 0.90. CONCLUSIONS Whole-tumor histogram analysis of DKI, IVIM, and DCE-MRI offers a noninvasive method for predicting the efficacy of c-KIT inhibitors in sinonasal MMs, presenting valuable implications for guiding targeted treatment in this rare cancer type.
Collapse
Affiliation(s)
- Cong Wang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Nuclear Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xuewei Niu
- Department of Nuclear Medicine, Hebei Medical University, Shijiazhuang, China
| | - Tianyi Xia
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Peng Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuzhe Wang
- Department of Radiology, Zhongshan Hospital of Fudan University, Fudan University, Shanghai, China
| | | | - Jianyuan Zhang
- Department of Nuclear Medicine, Baoding No. 1 Central Hospital, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zebin Xiao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
56
|
Steinberg FT, Simon M, Wawer-Matos Reimer PA, Rokohl AC, Heindl LM. [New systemic treatment approaches for conjunctival melanoma]. DIE OPHTHALMOLOGIE 2025; 122:349-356. [PMID: 40067450 DOI: 10.1007/s00347-025-02207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 02/05/2025] [Indexed: 05/08/2025]
Abstract
Conjunctival melanoma is a rare disease that nevertheless has a high tumor-associated mortality rate. A resection in sano with adjuvant local treatment currently represents the therapeutic gold standard and systemic treatment is used for metastasized conjunctival melanoma and/or very advanced nonresectable local findings. New knowledge on molecular changes in conjunctival melanoma shows a clear similarity to those of cutaneous melanoma. Therefore, many findings on new systemic forms of treatment for cutaneous melanoma can be transferred to conjunctival melanoma. In the clinical application BRAF/MEK inhibitors and immune checkpoint inhibitors are already in use and good response rates have been shown in small retrospective studies and case reports. Due to the rarity of conjunctival melanoma, there are no larger prospective studies comparing different systemic therapeutic agents. In a nonrandomized retrospective comparison, a better overall survival was shown for a combination of BRAF/MEK inhibitors (progression-free 1‑year survival probability of 54.7%; overall survival of 29.1 months) compared to a combination of PD1/CTLA4 antibodies (progression-free 1‑year survival probability of 42%; overall survival of 18 months). The current recommendation is to perform genomic profiling for every conjunctival melanoma, particularly to investigate a BRAF mutation. If a BRAF mutation is present, BRAF/MEK inhibitor treatment should preferably be initiated. Treatment with immune checkpoint inhibitors can be used in the case of BRAF-negative mutation status or treatment failure with BRAF/MEK inhibitors. Monotherapy with the CTLA4 antibody ipilimumab is not recommended due to its inferiority to PD1 antibodies. New knowledge in the genomic profiling of conjunctival melanoma could enable further targeted treatment options in the future.
Collapse
Affiliation(s)
| | - Michael Simon
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | | | - Alexander C Rokohl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| | - Ludwig M Heindl
- Zentrum für Augenheilkunde, Universitätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
57
|
Yuan M, Feng W, Ding H, Yang Y, Xu XS. Discovery of mutations predictive of survival benefit from immunotherapy in first-line NSCLC: A retrospective machine learning study of IMpower150 liquid biopsy data. Comput Biol Med 2025; 189:109964. [PMID: 40043417 DOI: 10.1016/j.compbiomed.2025.109964] [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: 01/30/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Predictive biomarker identification in cancer treatment has traditionally relied on pre-defined analyses, limiting discoveries to expected biomarkers and potentially overlooking novel ones predictive of therapy response. In this work, we develop a novel machine-learning approach capable of exploring full landscape of mutations and combinations and identify potentially new predictive biomarkers for chemoimmunotherapy. Utilizing the liquid biopsy dataset from 313 non-small cell lung cancer (NSCLC) patients in the Phase 3 Impower150 trial (NCT02366143), we developed the HRdiffRF algorithm with a novel hazard ratio-splitting criterion. Predictive mutations and combinations were identified for overall survival (OS) improvement with atezolizumab plus bevacizumab plus carboplatin and paclitaxel (ABCP) compared to bevacizumab plus carboplatin and paclitaxel (BCP). Our analysis confirms the predictive role of KRAS mutations and reveals the predictive value of PTPRD and SMARCA4 mutations in chemoimmunotherapy efficacy. Unlike other KRAS wild-type NSCLC patients, NSCLC patients with KRAS wild-type status and mutations in FAT1, ERBB2, or PTPRD may benefit from chemoimmunotherapy, while NTRK3 and GNAS mutations could negatively impact survival. Patients harboring concurrent KRAS and KEAP1 mutations may not benefit from chemoimmunotherapy. These findings highlight the complex genetic factors influencing treatment response for chemoimmunotherapy in NSCLC. In summary, the proposed machine-learning tool identified potential predictive biomarkers for first-line chemoimmunotherapy in NSCLC and can be readily applied to other tumor types and studies. It can also be extended to explore predictive biomarkers beyond mutations.
Collapse
Affiliation(s)
- Min Yuan
- Department of Health Data Science, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Wei Feng
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, China
| | - Haolun Ding
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, China
| | - Yaning Yang
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, China
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA.
| |
Collapse
|
58
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025; 25:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [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: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
59
|
Marranci A, Maresca L, Lodovichi S, Luserna di Rorà AG, Stecca B, Poliseno L. PARP1 in melanoma: Mechanistic insights and implications for basic and clinical research. Cancer Lett 2025; 617:217599. [PMID: 40024566 DOI: 10.1016/j.canlet.2025.217599] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma and have set a successful example for the treatment of other cancers. A similar breakthrough was achieved with the advent of PARP inhibitors (PARPi) in breast and ovarian cancer. Recent evidence highlights the critical role of PARP1 in melanoma initiation and progression. High PARP1 expression correlates with aggressive melanoma characteristics and poor patient outcomes. Preclinical and clinical data suggest that PARPi, alone or in combination, can effectively reduce melanoma cell viability and inhibit tumor growth. However, integrating PARPi with current treatment approaches and identifying patients who could benefit the most from such combinations remain underexplored areas of investigation. This review highlights the need for further basic and clinical research on PARP1 in melanoma, to better understand its role and to tackle major challenges in the field, such as resistance to targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Marranci
- Oncohematology Unit, Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, 56017, Pisa, Italy. http://www.fpscience.it/
| | - Luisa Maresca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, 50139, Florence, Italy
| | - Samuele Lodovichi
- Department of Biosciences, University of Milan, 20133, Milan, Italy; Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy
| | | | - Barbara Stecca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy; Oncogenomics Unit, Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 56124, Pisa, Italy.
| |
Collapse
|
60
|
Murad V, Metser U, Kohan A, Murad S, Veit-Haibach P, Ortega C. 18F-FDG PET/CT for the Detection of Immune-Related Adverse Events in Patients With Metastatic Melanoma Receiving Immunotherapy. Can Assoc Radiol J 2025:8465371251334929. [PMID: 40308072 DOI: 10.1177/08465371251334929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Purpose: To evaluate frequency and distribution of immune-related adverse events detected by 18F-FDG PET/CT in patients with metastatic melanoma undergoing immunotherapy. Materials and Methods: Retrospective observational cohort study evaluating 147 patients with metastatic melanoma treated with immunotherapy and referred for therapy response assessment with 18F-FDG PET/CT at our institution from January 2010 to August 2022. In total, 201 PET/CT scans performed at various time points were analyzed. IRAEs detected on PET/CT were compared against clinical reference standards, including physical examinations, laboratory tests, and biopsies. Diagnostic performance metrics (sensitivity, specificity, positive predictive value, negative predictive value), and diagnostic yields were calculated. Results: There were 36/147 patients (24.5%) with IRAEs recorded according to standard of reference, with 39 IRAEs in the entire cohort. At time point level, PET/CT identified 36/36 (100%) patients with IRAEs confirmed by the reference standard, while clinical suspicion identified 26/36 (72%) cases. At IRAE level, PET/CT identified 36/39 (92%) of IRAEs confirmed by the reference standard. Thirteen out of 39 (33.3%) cases identified on PET/CT were not suspected clinically but confirmed by the reference standard. The most frequent IRAEs, both suspected clinically and on PET/CT, corresponded to thyroiditis and colitis. Among the PET/CT positive cases, the majority corresponded to grade 2 severity. Conclusion: 18F-FDG PET/CT is highly effective in detecting IRAEs in patients with metastatic melanoma on immunotherapy, uncovering clinically unsuspected events in up to 33% of cases. These results highlight its important role in early detection, guiding timely interventions, and improving overall outcomes of immunotherapy-related toxicities.
Collapse
Affiliation(s)
- Vanessa Murad
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Ur Metser
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Andres Kohan
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Sarah Murad
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Patrick Veit-Haibach
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Claudia Ortega
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
61
|
Reitmajer M, Schäffeler N, Bach A, Nanz L, Amaral T, Leiter U, Flatz L, Forschner A. Psychosocial distress and persistent adverse events in long-term survivors of stage IV melanoma - a cross-sectional questionnaire study. J Dtsch Dermatol Ges 2025. [PMID: 40277327 DOI: 10.1111/ddg.15712] [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: 10/30/2024] [Accepted: 02/02/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors and targeted therapies have improved survival in patients with stage IV melanoma. However, the challenges faced by long-term survivors remain unclear. The long-term toxicity and psychosocial impact of these treatments in real-world patients have yet to be reported. MATERIAL AND METHODS We conducted a cross-sectional questionnaire study using established screening tools, including the Hornheide Screening Instrument (HSI), the Distress Thermometer (DT) with the National Comprehensive Cancer Network (NCCN) problem list, and melanoma-specific questions addressing persistent adverse events, social impairments, emotional needs, and financial concerns. RESULTS A total of 159 patients with stage IV melanoma (≥5 years after the initial diagnosis) were enrolled, of whom 93 completed the questionnaire. Approximately one-third of DT/HSI values exceeded the threshold, indicating a need for psycho-oncological support. More than 40% of patients reported persistent treatment-related complaints. Financial and work-related impacts were rare, affecting approximately 8% and 1% of patients, respectively. CONCLUSIONS High rates of psychosocial distress and persistent adverse events were observed, highlighting the need for cancer survivorship programs in the follow-up care of melanoma patients.
Collapse
Affiliation(s)
- Markus Reitmajer
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Norbert Schäffeler
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tuebingen, Tübingen, Germany
| | - Anne Bach
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tuebingen, Tübingen, Germany
| | - Lena Nanz
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
62
|
Xie Y, Wang R, Xu M, Chen J, Tan W, Chen Y, Bai Y, Wu N, Wu F, Xu X, Ma X, Liu Y. Potential of CLSPN as a therapeutic target in melanoma: a key player in melanoma progression and tumor microenvironment. J Transl Med 2025; 23:470. [PMID: 40275302 PMCID: PMC12020306 DOI: 10.1186/s12967-025-06455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Melanoma is a highly aggressive form of skin cancer. Despite significant advances in targeted therapies and immunotherapeutic approaches, some patients still have poor response rates, making a deeper understanding of melanoma pathogenesis essential. METHODS The expression of Claspin (CLSPN), prognosis and immune infiltration in skin cutaneous melanoma patients were analyzed by public databases. Immunohistochemistry was used to validate. Moreover, quantitative real-time polymerase chain reaction analysis, western blot, cell counting kit-8 assay, colony formation assay, flow cytometry, animal experiments, and RNA-seq were applied to explore its biological functions and potential molecular mechanisms of CLSPN in melanoma. RESULTS Our results demonstrated that abnormal CLSPN expression was correlated with poor prognosis in melanoma. Meanwhile, CLSPN may promote melanoma growth and progression in vivo and in vitro through IFI44L/JAK/STAT1 signaling. Additionally, CLSPN was associated with negative immune microenvironment in melanoma and may be related to polarization of tumor associated macrophages towards M2-type. CONCLUSIONS These findings suggest that CLSPN may be a promising new target for melanoma and accelerate personalized therapeutic strategies.
Collapse
Affiliation(s)
- Yongyi Xie
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Ruoqi Wang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Skin Disease Hospital, Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai, 200443, China
| | - Mingyuan Xu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Jiashe Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Wei Tan
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yanbin Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yun Bai
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Nanhui Wu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Fei Wu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiang Xu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China.
- , Baode Road 1278 street, Shanghai, 200433, China.
| | - Yeqiang Liu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Skin Disease Hospital, Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai, 200443, China.
- , Baode Road 1278 street, Shanghai, 200433, China.
| |
Collapse
|
63
|
Hanratty K, Finegan G, Rochfort KD, Kennedy S. Current Treatment of Uveal Melanoma. Cancers (Basel) 2025; 17:1403. [PMID: 40361330 PMCID: PMC12071000 DOI: 10.3390/cancers17091403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults worldwide [...].
Collapse
Affiliation(s)
- Katie Hanratty
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin 9, D09 V209 Dublin, Ireland; (K.H.); (G.F.); (K.D.R.)
- Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin 2, D02 XK51 Dublin, Ireland
| | - Gráinne Finegan
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin 9, D09 V209 Dublin, Ireland; (K.H.); (G.F.); (K.D.R.)
- Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin 2, D02 XK51 Dublin, Ireland
| | - Keith D. Rochfort
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin 9, D09 V209 Dublin, Ireland; (K.H.); (G.F.); (K.D.R.)
- Life Sciences Institute, Dublin City University, Collins Avenue, Glasnevin, Dublin 9, D09 V209 Dublin, Ireland
| | - Susan Kennedy
- Research Foundation, Royal Victoria Eye and Ear Hospital, Adelaide Road, Dublin 2, D02 XK51 Dublin, Ireland
- Life Sciences Institute, Dublin City University, Collins Avenue, Glasnevin, Dublin 9, D09 V209 Dublin, Ireland
| |
Collapse
|
64
|
Khaki AR, Reynolds KL, Petrillo LA. Beyond the Point: Learning From Outcomes After Inpatient Immune Checkpoint Inhibitor Therapy. JCO Oncol Pract 2025:OP2500213. [PMID: 40258200 DOI: 10.1200/op-25-00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Affiliation(s)
- Ali Raza Khaki
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Laura A Petrillo
- Harvard Medical School, Boston, MA
- Division of Palliative Care and Geriatrics, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
65
|
Hu M, Zhu X, Huang X, Hua L, Lin X, Zhang H, Hu Y, Tong T, Li L, Xuan B, Zhao Y, Zhou Y, Ding J, Ma Y, Jiang Y, Ning L, Zhang Y, Wang Z, Fang JY, Zhang Y, Xiao X, Hong J, Chen H, Li J, Chen H. Optimizing anti-PD-1/PD-L1 therapy efficacy and fecal microbiota transplantation donor selection through gut mycobiome-based enterotype. Cell Rep 2025; 44:115589. [PMID: 40257861 DOI: 10.1016/j.celrep.2025.115589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/06/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, but response variability remains a challenge. The gut microbiome's role in therapeutic efficacy is well established, but the impact of the gut mycobiome is less understood. Using unsupervised clustering, we identify two gut mycobiome-based enterotypes, favorable type and unfavorable type, characterized by distinct microbial compositions linked to immunotherapy outcomes. Favorable-type enterotypes exhibit higher fungal and bacterial alpha diversity, enriched butyrate-producing bacteria, and metabolic pathways related to butyric acid and sugar/starch metabolism. External validation confirms their predictive value in assessing immunotherapy efficacy. Multi-omics analysis reveals increased CD8+ T cell infiltration in the tumor microenvironment of favorable-type patients. Fecal microbiota transplantation (FMT) from favorable-type donors enhances anti-PD-1 sensitivity, promotes CD8+ T cell infiltration, and boosts butyrate production in vivo. These findings highlight the gut mycobiome's role in immunotherapy response and support FMT from favorable-type donors as a potential strategy for improving treatment outcomes and patient stratification.
Collapse
Affiliation(s)
- Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China; Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Li Hua
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaolin Lin
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ye Hu
- Department of Gastroenterology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Lingxi Li
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yi Jiang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Zhenyu Wang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou 221009, China
| | - Xiuying Xiao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Huimin Chen
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China.
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China.
| |
Collapse
|
66
|
Fager A, Samuelsson M, Olofsson Bagge R, Pivodic A, Bjursten S, Levin M, Jespersen H, Ny L. Immune checkpoint inhibitor therapy is associated with a decreased risk of developing melanoma brain metastases. BJC REPORTS 2025; 3:22. [PMID: 40217072 PMCID: PMC11992042 DOI: 10.1038/s44276-025-00137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 02/27/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Despite recent advancements in metastatic melanoma treatment, the emergence of melanoma brain metastases (MBM) continues to pose a challenge. This study aimed to explore factors associated with MBM development. METHODS This retrospective study included patients diagnosed with advanced melanoma (unresectable stages III and IV [M1a-c]) between 2013 and 2019 at Sahlgrenska University Hospital, Gothenburg, Sweden. Differences in baseline and primary tumor characteristics, mutational status, biomarker levels, and first-line treatment between patients who developed MBM (BM+) and patients who did not develop MBM (BM-) were analyzed using univariable and multivariable Cox proportional hazard regression. RESULT Of 395 patients, 91 subsequently developed MBM. Patients who received immune checkpoint inhibitors (ICI) as first-line treatment had a reduced risk of MBM development (p ≤ 0.001). None of the eleven patients who received CTLA-4 inhibitors as monotherapy or in combination with PD-1 inhibitors as first-line treatment developed brain metastases. Elevated plasma levels of S100B (p = 0.021) and higher metastatic stage (p = 0.047) were also associated with an increased risk of MBM development. CONCLUSION ICI treatment is associated with a decreased risk of MBM development, suggesting a protective role. Elevated S100B levels and stage IV disease at advanced melanoma diagnosis might indicate an increased risk of MBM development.
Collapse
Affiliation(s)
- Anna Fager
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Matilda Samuelsson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Sara Bjursten
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Max Levin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Lars Ny
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
67
|
Liu S, Liu J, Mei Y, Zhang W. Gut microbiota affects PD-L1 therapy and its mechanism in melanoma. Cancer Immunol Immunother 2025; 74:169. [PMID: 40214675 PMCID: PMC11992302 DOI: 10.1007/s00262-025-04018-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 blockade, have shown great success in treating melanoma. PD-L1 (B7-H1, CD274), a ligand of PD-1, binds to PD-1 on T cells, inhibiting their activation and proliferation through multiple pathways, thus dampening tumor-reactive T cell activity. Studies have linked PD-L1 expression in melanoma with tumor growth, invasion, and metastasis, making the PD-1/PD-L1 pathway a critical target in melanoma therapy. However, immune-related adverse events are common, reducing the effectiveness of anti-PD-L1 treatments. Recent evidence suggests that the gut microbiome significantly influences anti-tumor immunity, with the microbiome potentially reprogramming the tumor microenvironment and overcoming resistance to anti-PD-1 therapies in melanoma patients. This review explores the mechanisms of PD-1/PD-L1 in melanoma and examines how gut microbiota and its metabolites may help address resistance to anti-PD-1 therapy, offering new insights for improving melanoma treatment strategies.
Collapse
Affiliation(s)
- Shiqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jiahui Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yingwu Mei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Wenjuan Zhang
- Beijing Life Science Academy (BLSA), Beijing, China.
- Key Laboratory of Tobacco Flavor Basic Research of CNTC, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, China.
| |
Collapse
|
68
|
Rimassa L, Chan SL, Sangro B, Lau G, Kudo M, Reig M, Breder V, Ryu MH, Ostapenko Y, Sukeepaisarnjaroen W, Varela M, Tougeron D, Crysler OV, Bouattour M, Van Dao T, Tam VC, Faccio A, Furuse J, Jeng LB, Kang YK, Kelley RK, Paskow MJ, Ran D, Xynos I, Kurland JF, Negro A, Abou-Alfa GK. Five-year overall survival update from the HIMALAYA study of tremelimumab plus durvalumab in unresectable HCC. J Hepatol 2025:S0168-8278(25)00226-0. [PMID: 40222621 DOI: 10.1016/j.jhep.2025.03.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND & AIMS In the phase III HIMALAYA study (NCT03298451), STRIDE (Single Tremelimumab Regular Interval Durvalumab) significantly improved overall survival (OS) versus sorafenib in unresectable HCC (uHCC) and demonstrated long-term survival benefits. We report an updated exploratory analysis of OS with 5 years of follow-up, including survival by multiple tumour response measures. METHODS Participants were randomised to STRIDE (tremelimumab plus durvalumab), durvalumab or sorafenib. OS, depth of response and serious adverse events (AEs) were assessed. Extended long-term survivors (eLTS; ≥48 months beyond randomisation) were described. Updated data cut-off: 01 March 2024. RESULTS Median (95% CI) follow-up durations were 62.49 (59.47-64.79) months (STRIDE) and 59.86 (58.32-61.54) months (sorafenib). The OS HR (95% CI) for STRIDE versus sorafenib was 0.76 (0.65-0.89). OS rates at 60 months for STRIDE versus sorafenib were 19.6% versus 9.4% overall, 28.7% versus 12.7% in participants achieving disease control per RECIST v1.1 and 50.7% versus 26.3% in participants achieving >25% tumour shrinkage. No late-onset treatment-related serious AEs were reported for STRIDE. There were more eLTS with STRIDE (83/393, 21.1%) than sorafenib (45/389, 11.6%), and extended long-term survival occurred across all clinically relevant subgroups. CONCLUSIONS At 5 years, STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile. OS benefit with STRIDE was improved in participants with disease control. Data suggest that any degree of tumour shrinkage with STRIDE can be associated with improved OS, indicating that conventional response measures may not fully capture STRIDE benefits. Nevertheless, participants experiencing deep responses appear to have the greatest benefit. STRIDE continues to set new benchmarks in uHCC with 1 in 5 patients alive at 5 years. IMPACT AND IMPLICATIONS The phase III HIMALAYA study showed that STRIDE (Single Tremelimumab Regular Interval Durvalumab) improved overall survival (OS) versus sorafenib in participants with unresectable HCC (uHCC), including after 4 years of follow-up. Understanding the efficacy and safety of STRIDE over the longer term is important for healthcare providers; here, we demonstrate that STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile after 5 years of follow-up. OS benefit with STRIDE was improved in participants with disease control and any degree of tumour shrinkage, indicating that conventional response measures may not fully capture the benefits of STRIDE. These findings are important as they set new benchmarks in uHCC and may help guide clinical decisions in the future.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Yue-Kong Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra and CIBEREHD, Pamplona - Madrid, Spain
| | - George Lau
- Humanity and Health Clinical Trial Center, Humanity and Health Medical Group, Hong Kong SAR, China
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Maria Reig
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic de Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain
| | - Valeriy Breder
- Department of Chemotherapy, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yuriy Ostapenko
- Department of Minimally Invasive and Endoscopic Surgery, Interventional Radiology, National Cancer Institute, Kyiv, Ukraine
| | | | - Maria Varela
- Liver Unit, Hospital Universitario Central de Asturias, IUOPA, ISPA, FINBA, Universidad de Oviedo, Oviedo, Spain
| | - David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Oxana V Crysler
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Tu Van Dao
- Cancer Research and Clinical Trials Center, Department of Optimal Therapy, National Cancer Hospital, Hanoi, Vietnam
| | - Vincent C Tam
- Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Adilson Faccio
- Department of Oncology, CEON - Centro Especializado em Oncologia, Ribeirao Preto, Brazil
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Long-Bin Jeng
- Department of Surgery, China Medical University and Hospital, Taichung, Taiwan, Republic of China
| | - Yoon Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Michael J Paskow
- Global Medical Affairs, AstraZeneca, Gaithersburg, Maryland, USA
| | - Di Ran
- Statistics, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ioannis Xynos
- Oncology R&D, Late-Stage Development, AstraZeneca, Cambridge, UK
| | - John F Kurland
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Alejandra Negro
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Cornell University, New York, New York, USA; Weill Medical College, Cornell University, New York, New York, USA; Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
69
|
Casalegno Garduño R, Spitschak A, Pannek T, Pützer BM. CD8+ T Cell Subsets as Biomarkers for Predicting Checkpoint Therapy Outcomes in Cancer Immunotherapy. Biomedicines 2025; 13:930. [PMID: 40299510 PMCID: PMC12025007 DOI: 10.3390/biomedicines13040930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
The advent of immune checkpoint blockade (ICB) has transformed cancer immunotherapy, enabling remarkable long-term outcomes and improved survival, particularly with ICB combination treatments. However, clinical benefits remain confined to a subset of patients, and life-threatening immune-related adverse effects pose a significant challenge. This limited efficacy is attributed to cancer heterogeneity, which is mediated by ligand-receptor interactions, exosomes, secreted factors, and key transcription factors. Oncogenic regulators like E2F1 and MYC drive metastatic tumor environments and intertwine with immunoregulatory pathways, impairing T cell function and reducing immunotherapy effectiveness. To address these challenges, FDA-approved biomarkers, such as tumor mutational burden (TMB) and programmed cell death-ligand 1 (PD-L1) expression, help to identify patients most likely to benefit from ICB. Yet, current biomarkers have limitations, making treatment decisions difficult. Recently, T cells-the primary target of ICB-have emerged as promising biomarkers. This review explores the relationship between cancer drivers and immune response, and emphasizes the role of CD8+ T cells in predicting and monitoring ICB efficacy. Tumor-infiltrating CD8+ T cells correlate with positive clinical outcomes in many cancers, yet obtaining tumor tissue remains complex, limiting its practical use. Conversely, circulating T cell subsets are more accessible and have shown promise as predictive biomarkers. Specifically, memory and progenitor exhausted T cells are associated with favorable immunotherapy responses, while terminally exhausted T cells negatively correlate with ICB efficacy. Ultimately, combining biomarkers enhances predictive accuracy, as demonstrated by integrating TMB/PD-L1 expression with CD8+ T cell frequency. Computational models incorporating cancer and immune signatures could further refine patient stratification, advancing personalized immunotherapy.
Collapse
Affiliation(s)
- Rosaely Casalegno Garduño
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany; (R.C.G.); (A.S.); (T.P.)
| | - Alf Spitschak
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany; (R.C.G.); (A.S.); (T.P.)
| | - Tim Pannek
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany; (R.C.G.); (A.S.); (T.P.)
| | - Brigitte M. Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany; (R.C.G.); (A.S.); (T.P.)
- Department Life, Light & Matter, University of Rostock, 18059 Rostock, Germany
| |
Collapse
|
70
|
Bhojnagarwala PS, Jose J, Zhao S, Weiner DB. DNA-based immunotherapy for cancer: In vivo approaches for recalcitrant targets. Mol Ther 2025:S1525-0016(25)00282-5. [PMID: 40211538 DOI: 10.1016/j.ymthe.2025.04.008] [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: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/10/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment and complements traditional therapies, including surgery, chemotherapy, radiation therapy, and targeted therapies. Immunotherapy redirects the patient's immune system against tumors via several immune-mediated approaches. Over the past few years, therapeutic immunization, which enable the patient's T cells to better recognize and kill tumors, have been increasingly tested in the clinic, with several approaches demonstrating treatment improvements. There has been a renewed interest in cancer vaccines due to advances in tumor antigen identification, immune response optimization, novel adjuvants, next-generation vaccine delivery platforms, and antigen designs. The COVID-19 pandemic accelerated progress in nucleic acid-based vaccine manufacturing, which spurred broader interest in mRNA or plasmid platforms. Enhanced DNA vaccine designs, including optimized leader sequences and RNA and codon optimizations, improved formulations, and delivery via adaptive electroporation using stereotactic intramuscular/intradermal methods have improved T cell responses to plasmid-delivered tumor antigens. Additionally, advancements for direct in vivo delivery of DNA-encoded monospecific/bispecific antibodies offer novel tumor-targeting strategies. This review summarizes the recent clinical data for therapeutic cancer vaccines utilizing the DNA platform, including vaccines targeting common tumor-associated and viral antigens and neoantigen vaccines using nucleic acid technologies. We also summarize preclinical data using DNA-launched monoclonal/bispecific antibodies, underscoring their potential as a novel cancer therapy tool.
Collapse
Affiliation(s)
- Pratik S Bhojnagarwala
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Joshua Jose
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Shushu Zhao
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA.
| |
Collapse
|
71
|
Ye M, Ren S, Luo H, Wu X, Lian H, Cai X, Ji Y. Integration of graph neural networks and transcriptomics analysis identify key pathways and gene signature for immunotherapy response and prognosis of skin melanoma. BMC Cancer 2025; 25:648. [PMID: 40205338 PMCID: PMC11983817 DOI: 10.1186/s12885-025-13611-4] [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] [Accepted: 01/29/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVE The assessment of immunotherapy plays a pivotal role in the clinical management of skin melanoma. Graph neural networks (GNNs), alongside other deep learning algorithms and bioinformatics approaches, have demonstrated substantial promise in advancing cancer diagnosis and treatment strategies. METHODS GNNs models were developed to predict the response to immunotherapy and to pinpoint key pathways. Utilizing the genes from these key pathways, multi-omics bioinformatics methods were employed to refine the construction of a gene signature, termed responseScore, aimed at enhancing the precision of immunotherapy response predictions. Subsequently, responseScore was explored from the perspectives of prognosis, genetic variation, pathway enrichment, and the tumor microenvironment. Concurrently, the association among 13 genes contributing to responseScore and factors such as immunotherapy response, prognosis, and the tumor microenvironment was investigated. Among these genes, PSMB6 was subjected to an in-depth analysis of its biological effect through experimental approaches like transfection and co-culture. RESULTS In the finalized model utilizing GNNs, it has revealed an AUC of 0.854 within the training dataset and 0.824 within the testing set, pinpointing key pathways such as R-HSA-70,268. The indicator named as responseScore excelled in its predictive accuracy regarding immunotherapy response and patient prognosis. Investigations into genetic variation, pathway enrichment, tumor microenvironment disclosed a profound association between responseScore and the enhancement of immune cell infiltration and anti-tumor immunity. A negative correlation was observed between the expression of PSMB6 and immune genes, with elevated PSMB6 expression correlating with poor prognosis. ELISA detection after co-cultivation experiments revealed significant reductions in the levels of cytokines IL-6 and IL-1β in specimens from the PCDH-PSMB6 group. CONCLUSION The GNNs prediction model and the responseScore developed in this research effectively indicate the immunotherapy response and prognosis for patients with skin melanoma. Additionally, responseScore provides insights into the tumor microenvironment and the characteristics of tumor immunity of melanoma. Thirteen genes identified in this study show promise as potential tumor markers or therapeutic targets. Notably, PSMB6 emerges as a potential therapeutic target for skin melanoma, where its elevated expression exhibits an inhibitory effect on the tumor immunity.
Collapse
Affiliation(s)
- Maodong Ye
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Shuai Ren
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Huanjuan Luo
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Xiumin Wu
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Hongwei Lian
- Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Xiangna Cai
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China
| | - Yingchang Ji
- Medical Cosmetic Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China.
| |
Collapse
|
72
|
Sajan A, Badar W, Ahmed O. Transarterial Musculoskeletal Embolization of Immune Checkpoint Inhibitor-Induced Inflammatory Glenohumeral Joint Arthritis. J Vasc Interv Radiol 2025:S1051-0443(25)00280-5. [PMID: 40204183 DOI: 10.1016/j.jvir.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Affiliation(s)
- Abin Sajan
- Section of Vascular and Interventional Radiology, Department of Radiology, Columbia University Medical Center, New York, New York
| | - Wali Badar
- Section of Vascular and Interventional Radiology, Department of Radiology, University of Chicago Medicine, Chicago, Illinois
| | - Osman Ahmed
- Section of Vascular and Interventional Radiology, Department of Radiology, University of Chicago Medicine, Chicago, Illinois.
| |
Collapse
|
73
|
Adashek JJ, Moran JA, Le DT, Kurzrock R. Lessons learned from a decade of immune checkpoint inhibition: The good, the bad, and the ugly. Cancer Metastasis Rev 2025; 44:43. [PMID: 40183852 PMCID: PMC11971148 DOI: 10.1007/s10555-025-10260-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Discovering the brakes/checkpoints that cancer places on the immune system to prevent being eradicated led to the 2018 Nobel Prize and the development of multiple Food and Drug Administration-approved immune checkpoint inhibitors (ICIs). ICIs have transformed the treatment of numerous cancer types and, remarkably, some patients with end-stage metastatic disease can achieve durable, complete remissions - cures. Still, ICIs cause significant immune-related toxicities, and most tumors are resistant. Unusual progression patterns such as pseudo-progression and hyper-progression (accelerated progression) can occur. Biomarkers for ICI response/resistance include microsatellite instability, high tumor mutational burden, and PD-L1 immunohistochemistry positivity; but they are imperfect, perhaps because of immune system complexity. Herein, we explore the good, the bad, and the ugly of ICIs in cancer treatment.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Johns Hopkins Hospital, The Sidney Kimmel Comprehensive Cancer Center1800 Orleans St, Baltimore, MD, 21287, USA.
| | - Jillian A Moran
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Rd, Hanover, NH, 03755, USA
| | - Dung T Le
- Department of Oncology, The Johns Hopkins Hospital, The Sidney Kimmel Comprehensive Cancer Center1800 Orleans St, Baltimore, MD, 21287, USA
| | - Razelle Kurzrock
- WIN Consortium, 24 Rue Albert Thuret, 94550, Chevilly-Larue, Paris, France
- University of Nebraska, 6001 Dodge St, Omaha, NE, 68182, USA
- MCW Cancer Center, 8800 W Doyne Ave, Milwaukee, WI, 53226, USA
| |
Collapse
|
74
|
Oyama K, Nakata K, Abe T, Hirotaka K, Fujimori N, Kiyotani K, Iwamoto C, Ikenaga N, Morisaki S, Umebayashi M, Tanaka H, Koya N, Nakagawa S, Tsujimura K, Yoshimura S, Onishi H, Nakamura Y, Nakamura M, Morisaki T. Neoantigen peptide-pulsed dendritic cell vaccine therapy after surgical treatment of pancreatic cancer: a retrospective study. Front Immunol 2025; 16:1571182. [PMID: 40248703 PMCID: PMC12004129 DOI: 10.3389/fimmu.2025.1571182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Pancreatic cancer shows very poor prognosis and high resistance to conventional standard chemotherapy and immunotherapy; therefore, the development of new breakthrough therapies is highly desirable. Method We retrospectively evaluated the safety and efficacy of neoantigen peptide-pulsed dendritic cell (Neo-P DC) vaccine therapy after surgical treatment of pancreatic cancer. Result The result showed induction of neoantigen-specific T cells in 13 (81.3%) of the 16 patients who received Neo-P DC vaccines. In survival analysis of the nine patients who received Neo-P DC vaccines after recurrence, longer overall survival was observed in patients with neoantigen-specific T cell induction than those without T cell induction. Notably, only one of the seven patients who received Neo-P DC vaccines as adjuvant setting developed recurrence, and no patient died during median follow-up 61 months after surgery (range, 25-70 months). Furthermore, TCR repertoire analyses were performed in a case treated with Neo-P DC vaccine combined with long and short peptides, and one significantly dominant clone induced by the long peptide was detected among CD4+ T cell populations. Discussion The present study suggests the feasibility and efficacy of Neo-P DC vaccine therapy after surgical treatment of pancreatic cancer in both postoperative recurrence cases and adjuvant setting. A case analysis suggests the importance of combination with long peptides targeting CD4+ T cell.
Collapse
Affiliation(s)
- Koki Oyama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiya Abe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kento Hirotaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nao Fujimori
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuma Kiyotani
- Laboratory of Immunogenomics, Center for Intractable Diseases and ImmunoGenomics, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Morisaki
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayo Umebayashi
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Hiroto Tanaka
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Norihiro Koya
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Shinichiro Nakagawa
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Kenta Tsujimura
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Sachiko Yoshimura
- Corporate Headquarters, Cancer Precision Medicine Inc., Kawasaki, Japan
| | - Hideya Onishi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Nakamura
- Laboratory of Immunogenomics, Center for Intractable Diseases and ImmunoGenomics, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Morisaki
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| |
Collapse
|
75
|
Moura LIF, Malfanti A, Matos AI, Peres C, Armiñán A, Duro‐Castaño A, Conejos‐Sánchez I, Medel M, Đorđević S, Carrascosa P, Carreira B, Acúrcio RC, Xavier‐Ferreira H, Hernández‐Barranco A, Castellano E, Roselló E, Machado JC, Peinado H, Vicent MJ, Florindo HF. Off-The-Shelf Multivalent Nanoconjugate Cancer Vaccine Rescues Host Immune Response against Melanoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417348. [PMID: 39937158 PMCID: PMC12016742 DOI: 10.1002/adma.202417348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/18/2025] [Indexed: 02/13/2025]
Abstract
Tumor-associated antigen-based cancer vaccines suffer from limited clinical success compared to alternative immunotherapies in melanoma, an aggressive skin cancer with an immunosuppressive tumor microenvironment. The anti-tumor potential of a multivalent nanoconjugate cancer vaccine platform - a cross-linked star-shaped polyglutamate carrier (StCl) with marked lymphotropic character conjugated with melanoma-associated peptide antigens is evaluated through redox-responsive linkers. The co-delivery of melanoma-associated peptide antigens by the nanoconjugate platform induced significant effector immune responses in a mouse melanoma model. The nanoconjugate platform synergized with a PD-1 inhibitor to revert the immunosuppressive melanoma tumor microenvironment by improving cytotoxic T-cell infiltration, which prompted a superior anti-tumor effect with prolonged overall survival without acute organ toxicity. The antigen-specific anti-tumor immune response induced by the nanoconjugate platform is also validated in a melanoma patient-derived xenograft mouse model. A promising, versatile StCl-based platform is reported for generating off-the-shelf multivalent nanoconjugate cancer vaccines for the safe and efficient immunotherapeutic treatment of melanoma.
Collapse
Affiliation(s)
- Liane IF Moura
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
| | - Alessio Malfanti
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Present address:
Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia F. Marzolo 5Padova35131Italy
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
| | - Ana Armiñán
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
| | - Aroa Duro‐Castaño
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Present address:
CurapathAv. Benjamin Franklin 19Paterna46980Spain
| | - Inmaculada Conejos‐Sánchez
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
| | - María Medel
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
| | - Snežana Đorđević
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
- Present address:
Tosoh Bioscience GmbHIm Leuschnerpark 4, Griesheim64347HesseGermany
| | - Paula Carrascosa
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
| | - Bárbara Carreira
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
| | - Helena Xavier‐Ferreira
- i3S – Instituto de Investigação e Inovação em SaúdeMedical Faculty of PortoUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Alberto Hernández‐Barranco
- Microenvironment and Metastasis LaboratoryDepartment of Molecular OncologySpanish National Cancer Research Center (CNIO)C. de Melchor Fernández Almagro, 3Madrid28029Spain
| | - Elena Castellano
- Microenvironment and Metastasis LaboratoryDepartment of Molecular OncologySpanish National Cancer Research Center (CNIO)C. de Melchor Fernández Almagro, 3Madrid28029Spain
| | - Esther Roselló
- Department of PathologyHospital General Universitario de ValenciaAv. de les Tres Creus, 2, L'OliveretaValencia46014Spain
| | - José C. Machado
- i3S – Instituto de Investigação e Inovação em SaúdeMedical Faculty of PortoUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Héctor Peinado
- Microenvironment and Metastasis LaboratoryDepartment of Molecular OncologySpanish National Cancer Research Center (CNIO)C. de Melchor Fernández Almagro, 3Madrid28029Spain
| | - María J. Vicent
- Polymer Therapeutics LabPrince Felipe Research Center (CIPF)C/Eduardo Primo Yúfera 3Valencia46012Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa)Faculty of PharmacyUniversidade de LisboaAv. Prof. Gama PintoLisboa1649‐003Portugal
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Health Institute Carlos III (IISCIII)Av. Monforte de Lemos, 3‐5. Pabellón 11. Planta 0Madrid28029Spain
| |
Collapse
|
76
|
Murugan AK, Kannan S, Alzahrani AS. Immune checkpoint expression and therapeutic implications in IDH1-mutant and wild-type glioblastomas. Curr Probl Cancer 2025; 55:101182. [PMID: 39864140 DOI: 10.1016/j.currproblcancer.2025.101182] [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: 07/22/2024] [Revised: 11/17/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Programmed cell death protein 1 (PDCD1) and cluster of differentiation 274 (CD274) expression is implicated in escaping tumors from immune surveillance. Immune checkpoint inhibitors show promise in cancer therapy, yet their efficacy in glioblastomas, particularly with IDH1 mutations, remains unclear. This study analyzed two independent NGS datasets (n = 577 and n = 153) from TCGA to investigate the expression of PDCD1 and CD274 in glioblastomas and their relationship with IDH1 mutations. We used cBioPortal for mutation analysis, RNA seq for expression analysis, miRDB and miRabel for differential expression of miRNAs, and Kaplan-Meier for survival prediction. We found that 5.4% of glioblastomas harbored IDH1 mutations, correlating with improved overall survival (OS) (p = 2.196e-3). Different glioblastoma cohorts showed a diverse IDH1 mutational prevalence (4-31%). Despite this, IDH1Mu was consistently associated with better OS (p = 8.235e-5). Notably, PDCD1 and CD274 were statistically significantly highly expressed in both IDH1Wt (p < 0.0001) and IDH1Mu tumors (p < 0.0001), with higher expression linked to poorer survival outcomes (PDCD1: p = 0.009; CD274: p = 0.02). Differential co-expression analyses revealed distinct gene and miRNA profiles for IDH1Wt and IDH1Mu glioblastomas, with specific upregulation of PTEN and downregulation of MUC16 in IDH1Wt, and upregulation of PIK3R1 in IDH1Mu. Additionally, PIK3R1 and ITGB2 emerged as critical druggable targets. Our findings indicate that PDCD1 and CD274 are highly expressed irrespective of IDH1 mutation statuses, suggesting that glioblastomas could benefit from immunotherapy. Moreover, IDH1Mu glioblastomas may require a combination of PI3K/AKT/mTOR inhibitors and immunotherapy due to PIK3R1 overexpression.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia
| |
Collapse
|
77
|
de Groot S, Blommestein HM, Leeneman B, Uyl-de Groot CA, Haanen JBAG, Wouters MWJM, Aarts MJB, van den Berkmortel FWPJ, Blokx WAM, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Hospers GAP, Kapiteijn E, van Not OJ, van der Veldt AAM, Suijkerbuijk KPM, van Baal PHM. Development of a Decision Model to Estimate the Outcomes of Treatment Sequences in Advanced Melanoma. Med Decis Making 2025; 45:302-317. [PMID: 39985400 PMCID: PMC11894896 DOI: 10.1177/0272989x251319338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/17/2024] [Indexed: 02/24/2025]
Abstract
BackgroundA decision model for patients with advanced melanoma to estimate outcomes of a wide range of treatment sequences is lacking.ObjectivesTo develop a decision model for advanced melanoma to estimate outcomes of treatment sequences in clinical practice with the aim of supporting decision making. The article focuses on methodology and long-term health benefits.MethodsA semi-Markov model with a lifetime horizon was developed. Transitions describing disease progression, time to next treatment, and mortality were estimated from real-world data (RWD) as a function of time since starting treatment or disease progression and patient characteristics. Transitions were estimated separately for melanoma with and without a BRAF mutation and for patients with favorable and intermediate prognostic factors. All transitions can be adjusted using relative effectiveness of treatments derived from a network meta-analysis of randomized controlled trials (RCTs). The duration of treatment effect can be adjusted to obtain outcomes under different assumptions.ResultsThe model distinguishes 3 lines of systemic treatment for melanoma with a BRAF mutation and 2 lines of systemic treatment for melanoma without a BRAF mutation. Life expectancy ranged from 7.8 to 12.0 years in patients with favorable prognostic factors and from 5.1 to 8.7 years in patients with intermediate prognostic factors when treated with sequences consisting of targeted therapies and immunotherapies. Scenario analyses illustrate how estimates of life expectancy depend on the duration of treatment effect.ConclusionThe model is flexible because it can accommodate different treatments and treatment sequences, and the duration of treatment effects and the transitions influenced by treatment can be adjusted. We show how using RWD and data from RCTs can harness advantages of both data sources, guiding the development of future decision models.HighlightsThe model is flexible because it can accommodate different treatments and treatment sequences, and the duration of treatment effects as well as the transitions that are influenced by treatment can be adjusted.The long-term health benefits of treatment sequences depend on the place of different therapies within a treatment sequence.Assumptions about the duration of relative treatment effects influence the estimates of long-term health benefits.We show how the use of real-world data and data from randomized controlled trials harness the advantages of both data sources, guiding the development of future decision models.
Collapse
Affiliation(s)
- Saskia de Groot
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Hedwig M. Blommestein
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Brenda Leeneman
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - Carin A. Uyl-de Groot
- Institute for Medical Technology Assessment (iMTA), Erasmus University Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| | - John B. A. G. Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel W. J. M. Wouters
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, The Netherlands
- Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Maureen J. B. Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Willeke A. M. Blokx
- Department of Pathology, Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marye J. Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alfons J. M. van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Geke A. P. Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Olivier J. van Not
- Scientific Bureau, Dutch Institute for Clinical Auditing, Leiden, The Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Pieter H. M. van Baal
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, The Netherlands
| |
Collapse
|
78
|
Esmaeli B, Ogden T, Nichols M, Lu T, Matthew Debnam J, Dimitriou F, McQuade J, Glitza Oliva IC. Rate of response to immune checkpoint inhibitor therapy in patients with conjunctival melanoma. Melanoma Res 2025; 35:130-144. [PMID: 39656585 PMCID: PMC11894759 DOI: 10.1097/cmr.0000000000001016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Our primary objective was to estimate the overall response rate to immune checkpoint inhibitors (ICIs) in patients with locally advanced, multiply recurrent, or metastatic conjunctival melanoma treated with ICIs. A retrospective review of all consecutive conjunctival melanoma patients who were treated with ICI between October 2017 and January 2024 was carried out. The study included 16 patients with a median age of 66 years. The indications for ICI were locally extensive conjunctival melanoma in the eye/orbital area without nodal or distant metastasis in 10 patients, local recurrence of conjunctival melanoma and simultaneous nodal or distant metastasis in four patients, and metastatic conjunctival melanoma without local recurrence in two patients. Five patients received PD-1 inhibitor monotherapy with nivolumab or pembrolizumab; the other 11 received ipilimumab (CTLA-4 inhibitor) and nivolumab for several cycles and were then continued on nivolumab monotherapy ( n = 6) or not given additional ICI therapy ( n = 3). The number of cycles of ICI ranged from 2 to 25 (median, 13). Eight patients achieved a complete response. Six patients had progressive disease. The overall rate of objective response to ICI therapy was 63% (10 of 16), and for the subset of patients with local disease only, the objective response rate was 70% (7 of 10). In 14 patients (88%), orbital exenteration or additional extensive surgery was avoided; two patients had progression despite ICI and eventually needed an orbital exenteration. Future studies should aim to correlate biomarker data with response to ICI therapy in patients with conjunctival melanoma.
Collapse
Affiliation(s)
- Bita Esmaeli
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tyler Ogden
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Ophthalmology, Brooke Army Medical Center, San Antonio, Texas, USA
| | - Matthew Nichols
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Ophthalmology, Brooke Army Medical Center, San Antonio, Texas, USA
| | - Tracy Lu
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J. Matthew Debnam
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Florentia Dimitriou
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella C. Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
79
|
Floudas CS, Goswami M, Donahue RN, Pastor DM, Redman JM, Brownell I, Turkbey EB, Cordes LM, Steinberg SM, Manu M, Francis DC, Lamping E, Marté JL, Kackley M, Krauss E, Manukyan M, Jochems C, Schlom J, Gulley JL, Strauss J. Novel Combination Immunotherapy and Clinical Activity in Patients With HPV-Associated Cancers: A Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:394-399. [PMID: 39976981 PMCID: PMC11843463 DOI: 10.1001/jamaoncol.2024.6998] [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: 08/15/2024] [Accepted: 12/20/2024] [Indexed: 02/23/2025]
Abstract
IMPORTANCE Patients who experience progression of advanced human papillomavirus (HPV)-associated cancers and who have previously received first-line systemic treatment have a poor prognosis and limited therapeutic options. OBJECTIVE To assess the clinical activity of the combination of the HPV type 16 therapeutic vaccine PDS0101, the tumor-targeting interleukin 12 antibody-drug conjugate PDS01ADC, and the bifunctional anti-programmed cell death ligand 1 (PD-L1)/transforming growth factor β (TGF-β) bintrafusp alfa in advanced HPV-associated cancers. DESIGN, Setting, and Participants This nonrandomized clinical trial was phase 1/2 and investigator initiated, and was conducted at a single US cancer research center between June 2020 and July 2022. Patients with advanced or metastatic HPV-associated cancers were eligible, including patients who were both immune checkpoint blockade (ICB) naive and ICB resistant. The cutoff date for data analysis was May 13, 2024. INTERVENTION Patients received 1 mL of PDS0101 subcutaneously every 4 weeks for 6 doses then every 12 weeks for 2 additional doses, PDS01ADC, 16.8 µg/kg, subcutaneously every 4 weeks or PDS01ADC, 8 µg/kg, subcutaneously every 2 weeks, and bintrafusp alfa, 1200 mg, intravenously every 2 weeks. MAIN OUTCOMES AND MEASURES Objective response rate (ORR) by Response Evaluation Criteria in Solid Tumors version 1.1 in ICB-naive patients. RESULTS Of the 50 eligible patients, 26 (52%) were men and the median age was 56 years (range, 28-80 years). The median (IQR) follow-up was 37.7 (30.6-42.0) months. Fourteen patients (28%) were ICB naive, with an ORR of 35.7% (95% CI, 12.8%-64.9%), and median overall survival (OS) 42.4 months (95% CI, 8.3 months-not estimable); in ICB-resistant patients, the ORR was 16.7% (6 of 36 patients; 95% CI, 6.4%-32.8%) and median OS was 15.8 months (95% CI, 9.0-21.3 months). Among patients with HPV-16-positive tumors (37 patients [74%]), in the ICB-naive group (8 patients [21.6%]) the ORR was 62.5% (95% CI, 24.5%-91.5%) and a median OS measure was not reached. Grade 3 and 4 treatment-related adverse events occurred in 26 of 50 patients (52%). There were no treatment-related deaths. CONCLUSIONS AND RELEVANCE In this trial, the combination of PDS0101, PDS01ADC, and bintrafusp alfa showed an acceptable safety profile and promising antitumor activity and improved OS in patients with HPV-16-positive cancers, in both ICB-naive and ICB-resistant patients, warranting further evaluation of the combination of PDS0101 and PDS01ADC with simultaneous PD-L1/TGF-β inhibition in these populations. Trial Registration ClinicalTrials.gov Identifier: NCT04287868.
Collapse
Affiliation(s)
- Charalampos S. Floudas
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Meghali Goswami
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Renee N. Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Danielle M. Pastor
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason M. Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Isaac Brownell
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | - Evrim B. Turkbey
- Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa M. Cordes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michell Manu
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Deneise C. Francis
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Lamping
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L. Marté
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Kackley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Krauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Manuk Manukyan
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Caroline Jochems
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L. Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
80
|
Horisaki K, Yoshikawa S, Omata W, Tsutsumida A, Kiyohara Y. Comparison of efficacy and adverse events by treatment cycles of nivolumab and ipilimumab in Japanese melanoma patients: A single-center, retrospective study. J Dermatol 2025; 52:651-662. [PMID: 40040577 PMCID: PMC11975187 DOI: 10.1111/1346-8138.17672] [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: 12/05/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 03/06/2025]
Abstract
Combination therapy with nivolumab and ipilimumab (NIVO+IPI) is highly effective in treating advanced malignant melanoma (MM) but it is associated with a high incidence of treatment-related adverse events (TRAEs). This retrospective, cohort study evaluated the efficacy and TRAEs of NIVO+IPI in Japanese patients with unresectable stage III and IV MM, comparing outcomes based on the number of treatment cycles and the IPI dose. We reviewed data from 57 patients with advanced or recurrent MM who received NIVO+IPI at the Shizuoka Cancer Center between August 2015 and July 2024. Patients who received two or fewer NIVO + IPI cycles (NIVO+IPI ≤2 cycles) generally had worse Eastern Cooperative Oncology Group performance status and more advanced stages compared to those who received three or more cycles (NIVO+IPI ≥3 cycles). The analysis revealed that the NIVO+IPI ≥3 cycles group had significantly better overall survival compared to the NIVO+IPI ≤2 cycles group, although receiving three or more cycles was not an independent prognostic factor in multivariate analysis. There was no significant difference in the frequency or severity of TRAEs between the two groups, but the incidence of grade ≥3 TRAEs increased significantly between the first and second cycles of NIVO+IPI. Additionally, reducing the IPI dose from 3 mg/kg to 2 mg/kg appeared to lower the risk of grade ≥3 TRAEs. In conclusion, further research is needed to determine the optimal number of NIVO+IPI cycles for Japanese patients with advanced MM. However, assessing efficacy after the second cycle may help avoid unnecessary NIVO+IPI administration. Reducing the IPI dose to 2 mg/kg may also offer a safer treatment approach for these patients.
Collapse
Affiliation(s)
- Ken Horisaki
- Department of DermatologyShizuoka Cancer CenterShizuokaJapan
- Department of DermatologyNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Wataru Omata
- Department of DermatologyShizuoka Cancer CenterShizuokaJapan
| | | | - Yoshio Kiyohara
- Department of DermatologyShizuoka Cancer CenterShizuokaJapan
| |
Collapse
|
81
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [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: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
82
|
Nishimura Y, Estaris J, Koseki M, Elias E, Chesta F, Takaoka K, Shao T, Horita N, Fujiwara Y. Arthralgia in patients with cancer receiving immune checkpoint inhibitors: a systematic review and meta-analysis. Immunotherapy 2025; 17:437-446. [PMID: 40337868 DOI: 10.1080/1750743x.2025.2501519] [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: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Although immune checkpoint inhibitors (ICIs) are widely used for patients with cancer, evidence of the impact of ICIs on the incidence of arthralgia remains limited. OBJECTIVE To evaluate the impact of ICIs on arthralgia incidences in patients with cancer. METHODS We performed a systematic review to identify phase 3 randomized control trials (RCTs) evaluating ICIs in patients with cancer and reporting the incidence of arthralgia. We performed a meta-analysis to pool odds ratios (ORs) of any grade and grade 3-5 arthralgia. RESULTS Forty RCTs (n = 26,610) were included. The incidence of any-grade and grade 3-5 treatment-related arthralgia was 12.0% (n = 1,125/9,395) and 0.54% (n = 47/8,723). The addition of an ICI to systemic therapy, such as chemotherapy, significantly increased any-grade (OR 1.32, 95% CI: 1.13-1.54, p = 0.001) and grade 3-5 arthralgia (OR 1.78, 95% CI: 1.08-2.94, p = 0.02) with low heterogeneity among ICI subtype subgroups (I2 = 0%). ICI monotherapy was associated with higher incidences of arthralgia than non-taxane (OR 6.83, 95% CI: 3.05-15.30, p < 0.001) but not than taxane chemotherapy (OR 0.74, 95% CI: 0.44-1.24, p = 0.25). CONCLUSIONS These results could guide oncologists to assess arthralgia in patients receiving ICIs.
Collapse
Affiliation(s)
| | - Jonathan Estaris
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, USA
| | - Mako Koseki
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evelyn Elias
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fnu Chesta
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, USA
| | - Kensuke Takaoka
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, USA
| | - Theresa Shao
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nobuyuki Horita
- Chemotherapy Center, Yokohama City University Hospital, Yokohama, Japan
| | - Yu Fujiwara
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
83
|
Davidson SJ, Teoh DGK, Dudek AZ, Vogel RI. Immunotherapy for treatment of female genital tract melanoma: National Cancer Database analysis. Melanoma Res 2025:00008390-990000000-00201. [PMID: 40170584 DOI: 10.1097/cmr.0000000000001036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Goal of this study was to examine the impact of immunotherapy on overall survival (OS) in patients with female genital tract melanoma (GTM). This retrospective cohort study utilized the National Cancer Database to identify individuals with invasive vulvar or vaginal melanoma diagnosed between 2004 and 2019. Kaplan-Meier plots and multivariate Cox regression were used to describe the impact of immunotherapy on OS and to examine predictors of OS among those who received immunotherapy for those with vulvar or vaginal melanoma. Of the 870 patients with vaginal melanoma, 23.6% received immunotherapy. Receiving immunotherapy for treatment of vaginal melanoma was associated with improved OS (median: 21.8 versus 18.9 months; P = 0.01); this association remained after adjustment for other prognostic factors [hazard ratio (HR), 0.77; 95% confidence interval (CI), 0.62-0.95; P = 0.01]. The survival advantage was more pronounced among those who did not receive primary surgical resection (median: 18.6 versus 12.2 months; P = 0.0009). Among 3123 patients with vulvar melanoma, 15.3% received immunotherapy. Receiving immunotherapy for treatment of vulvar melanoma was associated with an improvement in OS (median: 43.6 versus 57.7 months; P = 0.06; HR, 0.86; 95% CI, 0.74-1.00; P = 0.04). Survival benefit was more pronounced when restricted to patients with advanced or unknown stage disease (median OS, 31.6 versus 24.2 months; P = 0.002; adjusted HR, 0.74; 95% CI, 0.61-0.89; P = 0.002) and among the small subset who did not receive primary surgical resection (median: 19.8 versus 9.6 months; P = 0.0005). Immunotherapy was associated with improved OS in patients with female GTM, with some subsets particularly benefitting.
Collapse
Affiliation(s)
| | - Deanna G K Teoh
- Department of Obstetrics, Gynecology and Women's Health
- Masonic Cancer Center, University of Minnesota, Minneapolis
| | | | - Rachel I Vogel
- Department of Obstetrics, Gynecology and Women's Health
- Masonic Cancer Center, University of Minnesota, Minneapolis
| |
Collapse
|
84
|
O'Sullivan HM, Conroy M, Power DG, Bambury RM, O'Mahony D, Collins DC, O'Leary MJ, O'Reilly S. Immune Checkpoint Inhibitors and Palliative Care at the End of Life: An Irish Multicentre Retrospective Study. J Palliat Care 2025; 40:147-151. [PMID: 35129002 DOI: 10.1177/08258597221078391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Background and Objectives: Immune checkpoint inhibitors (ICIs) have less toxicity than standard chemotherapy and are now standard of care for many patients with advanced cancer. A manageable side effect profile and potential for durable responses may lead to aggressive care of the palliative patient. We sought to evaluate palliative care input and ICI use at the end of life at two Irish cancer centres. Methods: We identified deceased patients who received at least one dose of an ICI between first of January 2013 to 31st of December 2018. A retrospective electronic chart review was performed. Results: The electronic records of 102 patients were analysed. Fifty eight percent were male and the median age of diagnosis of advanced disease was 60 years (range 17-78). Median time from last dose of ICI to death was 57 days (range 8-574) and 20% of patients died within 30 days of last dose of ICI. Most patients, 92%, were referred to palliative care. The median time from palliative care referral to death was 64 days (range 1- 1010). In the last 30 days of life, 39% of patients attended the emergency department (ED) and 46% had at least one hospital admission. Late palliative care referrals, ≤3 months before death, were associated with hospitalisations in the last month of life (64% vs. 36%, P = .02). Timing of palliative care referral did not affect ICI prescribing at the end of life (P = 0.38). ICI use in the last 30 days of life was not associated with increased ED presentations or hospitalisations at the end of life. Patients who received ICI in the last month had a higher likelihood of in-hospital death (43% vs. 16%, P = 0.02). Conclusions: ICI within 30 days of death was associated with dying in hospital but did not lead to more hospitalisations and emergency department presentations. Early palliative care did not affect ICI use but reduced hospitalisations at the end of life.
Collapse
Affiliation(s)
- H M O'Sullivan
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - M Conroy
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - D G Power
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Department of Medical Oncology, Mercy University Hospital, Cork, Ireland
| | - R M Bambury
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - D O'Mahony
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - D C Collins
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - M J O'Leary
- Department of Palliative Medicine, Marymount University Hospital and Hospice, Cork, Ireland
- Department of Palliative Medicine, Cork University Hospital, Cork, Ireland
| | - S O'Reilly
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
- Department of Medical Oncology, Mercy University Hospital, Cork, Ireland
- Cork Cancer Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
85
|
Brohet RM, de Boer ECS, Mossink JM, van der Eerden JJN, Oostmeyer A, Idzerda LHW, Maring JG, Paardekooper GMRM, Beld M, Lijffijt F, Dille J, de Groot JWB. Using Real-World Data for Machine-Learning Algorithms to Predict the Treatment Response in Advanced Melanoma: A Pilot Study for Personalizing Cancer Care. JCO Clin Cancer Inform 2025; 9:e2400181. [PMID: 40184559 DOI: 10.1200/cci-24-00181] [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: 07/16/2024] [Revised: 01/05/2025] [Accepted: 02/20/2025] [Indexed: 04/06/2025] Open
Abstract
PURPOSE The use of real-world data (RWD) in oncology is becoming increasingly important for clinical decision making and tailoring treatment. Despite the significant success of targeted therapy and immunotherapy in advanced melanoma, substantial variability in clinical responses to these treatments emphasizes the need for personalized approaches to therapy. MATERIALS AND METHODS In this pilot study, 239 patients with melanoma were included to predict the response to both targeted therapies and immunotherapies. We used machine learning (ML) to incorporate RWD and applied explainable artificial intelligence (XAI) to explain the individual predictions. RESULTS We developed, validated, and compared four ML models to evaluate 2-year survival using RWD. Our research showed encouraging outcomes, achieving an AUC of more than 80% and an estimated accuracy of over 74% across the four ML models. The random forest model exhibited the highest performance in predicting 2-year survival with an AUC of 0.85. Local interpretable model-agnostic explanations was used to explain individual predictions and provide trust and insights into the clinical implications of the ML model. CONCLUSION With this proof-of-concept, we integrated RWD into predictive modeling using ML techniques to predict clinical outcomes and explore their potential implications for clinical decision making. The potential of XAI was demonstrated to enhance trust and improve the usability of the model in clinical settings. Further research, including foundation modeling and generative AI, will likely increase the predictive power of prognostic and predictive ML models in advanced melanoma.
Collapse
Affiliation(s)
- Richard M Brohet
- Division Data Science, Department of Innovation and Science, Isala, Zwolle, the Netherlands
| | | | - Joram M Mossink
- Division Data Science, Department of Innovation and Science, Isala, Zwolle, the Netherlands
| | | | - Alexander Oostmeyer
- Division Data Science, Department of Innovation and Science, Isala, Zwolle, the Netherlands
| | - Luuk H W Idzerda
- Division Data Science, Department of Innovation and Science, Isala, Zwolle, the Netherlands
| | | | | | - Michel Beld
- Department of Business Intelligence, Isala, Zwolle, the Netherlands
| | - Fiona Lijffijt
- Department of Medical Ethics & Legal Affairs, Isala, Zwolle, the Netherlands
| | - Joep Dille
- Department of Innovation and Science, Isala, Zwolle, the Netherlands
| | | |
Collapse
|
86
|
Angell CD, Sun SH, Lapurga G, Benner B, Quiroga D, Savardekar H, DiVincenzo MJ, Abood D, Stiff A, Duggan M, Handley D, Nagle E, Harrison Howard J, Shah H, Kendra KL, Carson WE. A comparison of myeloid-derived suppressor cell populations in patients with ulcerated vs non-ulcerated melanoma receiving immune checkpoint blockade. Melanoma Res 2025; 35:102-108. [PMID: 39883562 PMCID: PMC11867852 DOI: 10.1097/cmr.0000000000001023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) are expanded in cancer patients, have an intrinsic immunosuppressive function, and thus may play a role in resistance to immunotherapy. Ulceration of the melanoma primary is associated with more aggressive disease and is an independent prognostic factor for melanoma-specific survival. However, the underlying factors contributing to this more aggressive phenotype are not completely understood. The current study aims to correlate changes in circulating MDSC during immunotherapy in patients with ulcerated vs non-ulcerated melanoma primary tumors. Longitudinal changes in levels of circulating MDSCs were analyzed via flow cytometry in melanoma patients receiving immune checkpoint inhibitors (ICIs) and stratified by ulceration status. Following the initiation of therapy, the percentage of total MDSCs increased significantly in patients with both ulcerated ( P = 0.003) and non-ulcerated ( P < 0.001) tumors. When MDSCs were stratified by subset, the proportion of granulocytic MDSC (PMN-MDSC) decreased in patients with non-ulcerated tumors ( P = 0.023), while the proportion remained stable in patients with ulcerated tumors ( P = 0.121). The reduction in the proportion PMN-MDSC in non-ulcerated patients coincided with a statistically significant increase in the proportion of CD14 + /CD15 + MDSC ( P = 0.008), resulting in a greater proportion of CD14 + /CD15 + MDSC in non-ulcerated patients as compared to ulcerated melanoma patients following two infusions of ICIs (27.3 ± 19.2% vs 16.1 ± 19.2%; P = 0.008). The trajectories of the MDSC populations described here provide insight into the altered tumor microenvironment in ulcerated melanoma and highlight key changes in a cell population that could contribute to immunotherapy resistance.
Collapse
Affiliation(s)
- Colin D Angell
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Steven H Sun
- Department of Surgery, Division of Surgical Oncology, The Ohio State University, Columbus, Ohio
| | - Gabriella Lapurga
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Dionisia Quiroga
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | | | | | - David Abood
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Andrew Stiff
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Megan Duggan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Demond Handley
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Erin Nagle
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - J Harrison Howard
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Hiral Shah
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Kari L Kendra
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Department of Surgery, Division of Surgical Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
87
|
Koizumi S, Yamazaki N, Ichigozaki Y, Kitagawa H, Kiniwa Y, Sato S, Takai T, Doi R, Ito T, Yasuda M, Kuwatsuka Y, Maekawa T, Asai J, Miyagawa T, Matsushita S, Funakoshi T, Yamamoto Y, Inozume T, Kishi A, Takenouchi T, Kokubu H, Ito S, Umeda Y, Yamamoto Y, Ishizuki S, Iino S, Uchi H, Nakagawa T, Inafuku K, Haga T, Kaneko T, Nakagawa M, Kamiya H, Arima M, Hoashi T, Hiura A, Kanazawa N, Manabe K, Ishikawa M, Asagoe K, Iwasawa U, Kadono T, Hatta N, Minami S, Nakano E, Ogata D, Fukushima S, Uhara H, Nakama K, Nakamura Y. Adjuvant Anti-PD-1 Monotherapy Versus Observation for Stage III Acral Melanoma of the Sole: A Multicenter Retrospective Study in Japanese Patients. JCO Glob Oncol 2025; 11:e2400644. [PMID: 40184568 PMCID: PMC12004986 DOI: 10.1200/go-24-00644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/16/2025] [Accepted: 02/28/2025] [Indexed: 04/06/2025] Open
Abstract
PURPOSE Adjuvant anti-PD-1 (adj PD-1) antibodies are extensively used to improve survival in patients with resected melanoma. Clinical trials on adj PD-1 antibodies have revealed significant improvements in recurrence-free survival (RFS); however, few of these trials have included patients with acral melanoma (AM). METHODS Clinical data were retrospectively collected from Japanese patients who underwent resection of stage III sole AM between 2014 and 2021. Survival outcomes, including RFS, distant metastasis-free survival (DMFS), and overall survival (OS), were compared between patients without adjuvant therapy (OBS group) and those receiving adj PD-1 group. RESULTS This study included 139 patients (OBS: 79; adj PD-1: 60), with a median follow-up of 2.6 years. The baseline characteristics were comparable, except for age and nodal metastasis. No significant differences in survival were observed between the OBS and adj PD-1 groups (3-year RFS: 36.7% v 27.5%, P = .13; 3-year DMFS: 51.0% v 45.3%, P = .51; 3-year OS: 65.3% v 67.4%, P = .45). Multivariate analysis showed no survival benefit of adj PD-1 (RFS: hazard ratio [HR], 1.25, P = .29; DMFS: HR, 1.03, P = .89; and OS: HR, 0.69, P = .23). Each survival outcome after propensity score matching confirmed no significant difference between the matched OBS group (n = 52) and adj PD-1 group (n = 52; 3-year RFS: 34.3% v 25.9%, P = .22; 3-year DMFS: 45.6% v 46.5%, P = .85; 3-year OS: 60.7% v 68.9%, P = .29). CONCLUSION Adj PD-1 did not improve the prognosis in sole AM. However, further studies are essential to evaluate the efficacy of the adj anti-PD-1 antibody in AM.
Collapse
Affiliation(s)
- Shigeru Koizumi
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
- Department of Dermatology, Chiba University, Chiba, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Ichigozaki
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Yukiko Kiniwa
- Department of Dermatology, Shinshu University, Matsumoto, Japan
| | - Sayuri Sato
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiro Takai
- Department of Dermatology, Hyogo Cancer Center, Akashi, Japan
| | - Reiichi Doi
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Yutaka Kuwatsuka
- Department of Dermatology and Allergology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takeo Maekawa
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
- Department of Dermatology, Jichi Medical University, Saitama Medical Center, Saitama, Japan
| | - Jun Asai
- Department of Dermatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuya Miyagawa
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology, NHO Kagoshima Medical Center, Kagoshima, Japan
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | | | | | - Akiko Kishi
- Department of Dermatology, Toranomon Hospital, Tokyo, Japan
| | - Tatsuya Takenouchi
- Department of Dermatology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiraku Kokubu
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Shusaku Ito
- Department of Dermatology, Hitachi General Hospital, Hitachi, Japan
| | - Yoshiyasu Umeda
- Department of Dermatology, Kawasaki Medical School, Kurashiki, Japan
| | - Yuki Yamamoto
- Department of Dermatology, Wakayama Medical University, Wakayama, Japan
| | | | - Shiro Iino
- Department of Dermatology, University of Fukui, Fukui, Japan
| | - Hiroshi Uchi
- Department of Dermato-Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Tomoe Nakagawa
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Kazuhiro Inafuku
- Department of Dermatology, Kimitsu Chuo Hospital, Kisarazu, Japan
| | - Takahiro Haga
- Department of Dermatology, Kesennuma City Hospital, Miyagi, Japan
| | - Takahide Kaneko
- Department of Dermatology, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Masahiro Nakagawa
- Department of Plastic and Reconstructive Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Kamiya
- Department of Dermatology, Central Japan International Medical Center, Gifu, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Aichi, Japan
| | - Toshihiko Hoashi
- Department of Dermatology, Nippon Medical School Hospital, Tokyo, Japan
| | - Azusa Hiura
- Department of Dermatology, Teikyo University, Tokyo, Japan
| | - Nobuo Kanazawa
- Department of Dermatology, Hyogo Medical University, Hyogo, Japan
| | - Keiko Manabe
- Department of Dermatology, Takamatsu Red Cross Hospital, Takamatsu, Japan
| | - Masashi Ishikawa
- Department of Dermatology, Saitama Cancer Center, Saitama, Japan
| | - Kenji Asagoe
- Department of Dermatology, NHO Okayama Medical Center, Okayama, Japan
| | - Utsugi Iwasawa
- Department of Dermatology, Metropolitan Hiroo Hospital, Tokyo, Japan
| | - Takafumi Kadono
- Department of Dermatology, St Marianna University, Kawasaki, Japan
| | - Naohito Hatta
- Department of Dermatology, Toyama Prefectural Central Hospital, Toyama, Japan
| | | | - Eiji Nakano
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenta Nakama
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Yasuhiro Nakamura
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
88
|
Hill J, Schoenfeld JD. Immunotherapy With Curative Intent Radiotherapy for Patients With Cancers of the Head and Neck. Semin Radiat Oncol 2025; 35:214-223. [PMID: 40090748 DOI: 10.1016/j.semradonc.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 03/18/2025]
Affiliation(s)
- Jordan Hill
- Banner MD Anderson Cancer Center, Gilbert, AZ.
| | | |
Collapse
|
89
|
Ma K, Xu Y, Cheng H, Tang K, Ma J, Huang B. T cell-based cancer immunotherapy: opportunities and challenges. Sci Bull (Beijing) 2025:S2095-9273(25)00337-8. [PMID: 40221316 DOI: 10.1016/j.scib.2025.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/24/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
T cells play a central role in the cancer immunity cycle. The therapeutic outcomes of T cell-based intervention strategies are determined by multiple factors at various stages of the cycle. Here, we summarize and discuss recent advances in T cell immunotherapy and potential barriers to it within the framework of the cancer immunity cycle, including T-cell recognition of tumor antigens for activation, T cell trafficking and infiltration into tumors, and killing of target cells. Moreover, we discuss the key factors influencing T cell differentiation and functionality, including TCR stimulation, costimulatory signals, cytokines, metabolic reprogramming, and mechanistic forces. We also highlight the key transcription factors dictating T cell differentiation and discuss how metabolic circuits and specific metabolites shape the epigenetic program of tumor-infiltrating T cells. We conclude that a better understanding of T cell fate decision will help design novel strategies to overcome the barriers to effective cancer immunity.
Collapse
Affiliation(s)
- Kaili Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Yingxi Xu
- Department of Oncology, University of Lausanne, Lausanne, 1015, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, 1066, Switzerland; National Key Laboratory of Blood Science, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 300070, China
| | - Hongcheng Cheng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Ke Tang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bo Huang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
90
|
Lim SY, Boyd SC, Diefenbach RJ, Rizos H. Circulating MicroRNAs: functional biomarkers for melanoma prognosis and treatment. Mol Cancer 2025; 24:99. [PMID: 40156012 PMCID: PMC11951542 DOI: 10.1186/s12943-025-02298-7] [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: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
MicroRNAs (miRNAs) hold significant promise as circulating cancer biomarkers and unlike many other molecular markers, they can provide valuable insights that extend beyond tumour biology. The expression of circulating miRNAs may parallel the cellular composition and dynamic activity within the tumour microenvironment and reveal systemic immune responses. The functional complexity of miRNAs-where a single miRNA can regulate multiple messenger RNAs (mRNAs) to fine tune fundamental processes, and a single mRNA can be targeted by multiple miRNAs-underscores their broad significance and impact. However, this complexity poses significant challenges for translating miRNA research into clinical practice. In melanoma, specific miRNA signatures have shown notable diagnostic, prognostic and predictive value, with lineage-specific and immune-related miRNAs frequently identified as valuable markers. In this review, we explore the role of circulating miRNAs as potential biomarkers in melanoma, and highlight the current status and advances required to translate miRNA research into therapeutic opportunities.
Collapse
Affiliation(s)
- Su Yin Lim
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Suzanah C Boyd
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Russell J Diefenbach
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Helen Rizos
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
91
|
Middha P, Thummalapalli R, Quandt Z, Balaratnam K, Cardenas E, Falcon CJ, Margaret Lung Group P, Gubens MA, Huntsman S, Khan K, Li M, Lovly CM, Patel D, Zhan LJ, Liu G, Aldrich MC, Schoenfeld A, Ziv E. Germline prediction of immune checkpoint inhibitor discontinuation for immune-related adverse events. J Immunother Cancer 2025; 13:e011273. [PMID: 40154961 PMCID: PMC11956315 DOI: 10.1136/jitc-2024-011273] [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: 12/10/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) can yield remarkable clinical responses in subsets of patients with solid tumors, but they also commonly cause immune-related adverse events (irAEs). The predictive features of clinically severe irAEs leading to cessation of ICIs have yet to be established. Given the similarities between irAEs and autoimmune diseases, we sought to investigate the association of a germline polygenic risk score for autoimmune disease and discontinuation of ICIs due to irAEs. METHODS The Genetics of immune-related adverse events and Response to Immunotherapy (GeRI) cohort comprises 1302 patients with non-small cell lung cancer (NSCLC) who received ICI therapy between 2009 and 2022 at four academic medical centers. We used a published polygenic risk score for autoimmune diseases (PRSAD) in the general population and validated it in the All of Us. We then assessed the association between PRSAD and cessation of ICI therapy due to irAEs in the GeRI cohort, using cause-specific and Fine-Gray subdistribution hazard models. To further understand the differential effects of type of therapy on the association between PRSAD and cessation of ICI due to irAEs, we conducted a stratified analysis by type of ICI therapy. RESULTS Using a competing risk model, we found an association between PRSAD and ICI cessation due to irAEs (HR per SD=1.24, p=0.004). This association was particularly strong in patients who had ICI cessation due to irAEs within 3 months of therapy initiation (HR per SD=1.40, p=0.005). Individuals in the top quintile of PRSAD had 4.8% ICI discontinuation for irAEs by 3 months, compared with 2% discontinuation by 3 months among patients in the bottom quintile (log-rank p=0.03). In addition, among patients who received combination programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte associated protein 4 (CTLA4) inhibitors, ICI discontinuation for irAEs by 3 months occurred in 4 of the 13 patients (30.8%) with high PRSAD genetic risk (top quintile) versus 3 of 21 patients (14.3%) with low PRSAD genetic risk (bottom quintile). CONCLUSIONS We demonstrate an association between a polygenic risk score for autoimmune disease and early ICI discontinuation for irAEs. Our results suggest that germline genetics may be used as an adjunctive tool for risk stratification around ICI clinical decision-making in solid tumor oncology.
Collapse
Affiliation(s)
- Pooja Middha
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Rohit Thummalapalli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zoe Quandt
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| | | | - Eduardo Cardenas
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Christina J Falcon
- Fiona and Stanley Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Matthew A Gubens
- Division of Hematology and Oncology and UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Scott Huntsman
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Khaleeq Khan
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Min Li
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Christine M Lovly
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
| | - Devalben Patel
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Luna Jia Zhan
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Temerty School of Medicine, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adam Schoenfeld
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, Center for Genes, Environment and Health and Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
92
|
Berry D, Moldoveanu D, Rajkumar S, Lajoie M, Lin T, Tchelougou D, Sakthivel S, Sharon I, Bernard A, Pelletier S, Ripstein Y, Spatz A, Miller WH, Jamal R, Lapointe R, Mes-Masson AM, Petrecca K, Meguerditchian AN, Richardson K, Wang B, Chergui M, Guiot MC, Watters K, Stagg J, Schmeing TM, Rodier F, Turcotte S, Mihalcioiu C, Meterissian S, Watson IR. The NF1 tumor suppressor regulates PD-L1 and immune evasion in melanoma. Cell Rep 2025; 44:115365. [PMID: 40023845 DOI: 10.1016/j.celrep.2025.115365] [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: 10/20/2023] [Revised: 11/24/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Hotspot BRAF, hotspot NRAS, and NF1 loss-of-function mutations are found in approximately 50%, 25%, and 15% of cutaneous melanomas, respectively. Compared to mutant BRAF and NRAS, the role of NF1 loss in melanoma remains understudied. NF1 has a RAS GTPase-activating protein (GAP) function; however, studies also support NF1 RAS-independent tumor-suppressor functions. Recent reports indicate that patients with NF1 mutant melanoma have high response rates to anti-PD-1 immune checkpoint inhibitors (ICIs) for reasons that are not entirely clear. Here, we present data demonstrating that NF1 interacts with PD-L1. Furthermore, NF1 loss in melanoma lines increases PD-L1 cell surface expression through a RAS-GAP-independent mechanism. Co-culture experiments demonstrate that NF1 depletion in melanoma increases resistance to T cell killing, which can be abrogated with anti-PD-1/PD-L1 ICIs. These results support a model whereby NF1 loss leads to immune evasion through the PD-L1/PD-1 axis, providing support for the examination of anti-PD-1 therapies in other NF1 mutant cancers.
Collapse
Affiliation(s)
- Diana Berry
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Dan Moldoveanu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Mathieu Lajoie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Tiffany Lin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Daméhan Tchelougou
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Samridhi Sakthivel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Antoine Bernard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Sandy Pelletier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Yael Ripstein
- Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Alan Spatz
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Wilson H Miller
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Rahima Jamal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | | | | | - Beatrice Wang
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - May Chergui
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | | | - Kevin Watters
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Simon Turcotte
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | | | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada.
| |
Collapse
|
93
|
Lemaître S, Arora AK, Hay GR, Sagoo MS. Spectrum of presentation of intraocular metastases from cutaneous melanoma in the era of immunotherapy and targeted therapies. Eye (Lond) 2025:10.1038/s41433-025-03753-x. [PMID: 40128380 DOI: 10.1038/s41433-025-03753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 10/29/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Intraocular metastases from cutaneous melanoma are rare. Diagnosis can be challenging and there is currently no consensus on treatment. However, with the increasing incidence of this cancer and improved survival of patients treated with targeted BRAF-MEK inhibitors and checkpoint inhibitors, it is likely that more cases will be referred to ocular oncology clinics. SUBJECTS Single-centre retrospective study. We included all the patients diagnosed with intraocular metastases from cutaneous melanoma seen between 2017 and 2022. RESULTS The first patient had bilateral choroidal metastases and unilateral vitreous cells (treated with external beam radiotherapy and immunotherapy), the second had unilateral amelanotic vitreous metastasis (treated with vitrectomy and BRAF-MEK inhibitors) and the third had bilateral multifocal choroidal metastases (treated with BRAK-MEK inhibitors followed by immunotherapy). The fourth patient (previously reported) had unilateral anterior segment and vitreous metastases (treated with immunotherapy and enucleation). Interestingly, two patients had a history of uveitis in the affected eye, unrelated to the ocular metastases. All four patients had synchronous systemic metastases. CONCLUSIONS The diagnosis of intraocular metastases from cutaneous melanoma is generally clinical but it is sometimes challenging because of possible masquerade syndromes. The presence of other extraocular metastatic sites is an indicator of the diagnosis. Cytopathologic proof combined with genetic analysis is sometimes necessary for diagnosis, especially with amelanotic vitreous debris or in rare cases where systemic screening is negative. New treatments with targeted BRAF-MEK inhibitors and checkpoint inhibition may avoid external beam radiotherapy and enucleation in some patients.
Collapse
Affiliation(s)
- Stéphanie Lemaître
- Service d'oculoplastique, Hôpital ophtalmique Jules-Gonin-Fondation Asile des aveugles, Lausanne, Switzerland.
- Moorfields Eye Hospital, London, UK.
| | | | | | - Mandeep S Sagoo
- Department of Ophthalmology, The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
94
|
Bennion KB, Miranda R.Bazzano J, Liu D, Wagener M, Paulos CM, Ford ML. Macrophage-derived Fgl2 dampens antitumor immunity through regulation of FcγRIIB+CD8+ T cells in melanoma. JCI Insight 2025; 10:e182563. [PMID: 40125553 PMCID: PMC11949062 DOI: 10.1172/jci.insight.182563] [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: 05/01/2024] [Accepted: 02/05/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer immunotherapy has emerged as a promising therapeutic modality but heterogeneity in patient responsiveness remains. Thus, greater understanding of the immunologic factors that dictate response to immunotherapy is critical to improve patient outcomes. Here, we show that fibrinogen-like protein 2 (Fgl2) is elevated in the setting of melanoma in humans and mice and plays a functional role in inhibiting the CD8+ T cell response. Surprisingly, the tumor itself is not the major cellular source of Fgl2. Instead, we found that macrophage-secreted Fgl2 dampens the CD8+ T cell response through binding and apoptosis of FcγRIIB+CD8+ T cells. This regulation was CD8+ T cell autonomous and not via an antigen-presenting cell intermediary, as absence of Fcgr2b from the CD8+ T cells rendered T cells insensitive to Fgl2 regulation. Fgl2 is robustly expressed by macrophages in 10 cancer types in humans and in 6 syngeneic tumor models in mice, underscoring the clinical relevance of Fgl2 as a therapeutic target to promote T cell activity and improve patient immunotherapeutic response.
Collapse
Affiliation(s)
- Kelsey B. Bennion
- Cancer Biology PhD program
- Department of Surgery
- Winship Cancer Institute
| | | | - Danya Liu
- Department of Surgery
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maylene Wagener
- Department of Surgery
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Mandy L. Ford
- Cancer Biology PhD program
- Department of Surgery
- Winship Cancer Institute
- Immunology and Molecular Pathogenesis PhD program, and
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
95
|
Garde C, Pavlidis MA, Garces P, Lange EJ, Ramarathinam SH, Sokač M, Pandey K, Faridi P, Ahrenfeldt J, Chung S, Friis S, Kleine-Kohlbrecher D, Birkbak NJ, Kringelum JV, Rønø B, Purcell AW, Trolle T. Endogenous viral elements constitute a complementary source of antigens for personalized cancer vaccines. NPJ Vaccines 2025; 10:54. [PMID: 40113807 PMCID: PMC11926357 DOI: 10.1038/s41541-025-01107-y] [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: 10/03/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Personalized cancer vaccines (PCVs) largely leverage neoantigens arising from somatic mutations, limiting their application to patients with relatively high tumor mutational burden (TMB). This underscores the need for alternative antigens to design PCVs for low TMB cancers. To this end, we substantiate endogenous retroviral elements (EVEs) as tumor antigens through large-scale genomic analyses of healthy tissues and solid cancers. These analyses revealed that the breadth of EVE expression in tumors stratify checkpoint inhibitor-treated melanoma patients into groups with differential overall and progression-free survival. To enable the design of PCVs containing EVE-derived epitopes with therapeutic potential, we developed a computational pipeline, ObsERV. We show that EVE-derived peptides are presented as epitopes on tumors and can be predicted by ObsERV. Preclinical testing of ObsERV demonstrates induction of sustained poly-functional CD4+ and CD8+ T-cell responses as well as long-term tumor protection. As such, EVEs may facilitate and improve PCVs, especially for low-TMB patients.
Collapse
Affiliation(s)
- Christian Garde
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark.
| | | | - Pablo Garces
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Emma J Lange
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Sri H Ramarathinam
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mateo Sokač
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kirti Pandey
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Shanzou Chung
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stine Friis
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | | | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Birgitte Rønø
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Thomas Trolle
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| |
Collapse
|
96
|
Santibanez JF. Myeloid-Derived Suppressor Cells: Implications in Cancer Immunology and Immunotherapy. FRONT BIOSCI-LANDMRK 2025; 30:25203. [PMID: 40152373 DOI: 10.31083/fbl25203] [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: 06/11/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 03/29/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) are believed to be key promoters of tumor development and are recognized as a hallmark of cancer cells' ability to evade the immune system evasion. MDSC levels often increase in peripheral blood and the tumor microenvironment (TME). These cells exert immunosuppressive functions, weakening the anticancer immune surveillance system, in part by repressing T-cell immunity. Moreover, MDSCs may promote tumor progression and interact with cancer cells, increasing MDSC expansion and favoring an immunotolerant TME. This review analyzes the primary roles of MDSCs in cancer and T-cell immunity, discusses the urgent need to develop effective MDSC-targeted therapies, and highlights the potential synergistic combination of MDSC targeting with chimeric antigen receptors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia
- Integrative Center for Biology and Applied Chemistry (CIBQA), Bernardo O'Higgins University, 8370993 Santiago, Chile
| |
Collapse
|
97
|
Wang W, Li X, Hu R, Dong L, Pei S, Jin L, Gao Q, Chen X, Yin M. BET inhibitor in combination with BCG vaccine enhances antitumor efficacy and orchestrates T cell reprogramming for melanoma. Cell Rep Med 2025; 6:101995. [PMID: 40107246 PMCID: PMC11970395 DOI: 10.1016/j.xcrm.2025.101995] [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: 08/07/2023] [Revised: 08/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
Immunotherapy shows remarkable benefits in treating melanoma, yet existing approaches achieve limited overall responses. Here, we show that a combination of bromodomain and extra-terminal protein family inhibitor, NHWD-870, and Bacillus Calmette-Guérin vaccine is a promising therapeutic strategy for melanomas. Single-cell transcriptome analyses and functional experiments show that the combination therapy significantly inhibited tumor growth by reprogramming T cells toward an immune-activated state, enhancing their cytotoxicity, preventing their exhaustion, and increasing the recruitment of them into the tumor microenvironment. We identify the molecule, MT1, as a direct downstream target of BRD4, which is effectively suppressed by NHWD-870. Furthermore, our findings are reinforced by a humanized patient-derived xenograft (PDX) model, which exhibits notable antitumor effects in humanized tumor-bearing mice treated with the combination therapy. Our study underscores the immense potential of this therapeutic approach for clinical practice, offering promising prospects in overcoming the limitations of current treatments.
Collapse
Affiliation(s)
- Wenhua Wang
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xin Li
- Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing 404100, China; Translational Medicine Research Center (TMRC), School of Medicine Chongqing University, Shapingba, Chongqing 400000, China.
| | - Rui Hu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Liang Dong
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Shiyao Pei
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Liping Jin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Qian Gao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China.
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China.
| |
Collapse
|
98
|
Agostini M, Traldi P, Hamdan M. Programmed Cell Death Ligand as a Biomarker for Response to Immunotherapy: Contribution of Mass Spectrometry-Based Analysis. Cancers (Basel) 2025; 17:1001. [PMID: 40149335 PMCID: PMC11940629 DOI: 10.3390/cancers17061001] [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: 02/08/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Immune checkpoint inhibition is a major component in today's cancer immunotherapy. In recent years, the FDA has approved a number of immune checkpoint inhibitors (ICIs) for the treatment of melanoma, non-small-cell lung, breast and gastrointestinal cancers. These inhibitors, which target cytotoxic T-lymphocyte antigen-4, programmed cell death (PD-1), and programmed cell death ligand (PD-L1) checkpoints have assumed a leading role in immunotherapy. The same inhibitors exert significant antitumor effects by overcoming tumor cell immune evasion and reversing T-cell exhaustion. The initial impact of this therapy in cancer treatment was justly described as revolutionary, however, clinical as well as research data which followed demonstrated that these innovative drugs are costly, are associated with potentially severe adverse effects, and only benefit a small subset of patients. These limitations encouraged enhanced research and clinical efforts to identify predictive biomarkers to stratify patients who are most likely to benefit from this form of therapy. The discovery and characterization of this class of biomarkers is pivotal in guiding individualized treatment against various forms of cancer. Currently, there are three FDA-approved predictive biomarkers, however, none of which on its own can deliver a reliable and precise response to immune therapy. Present literature identifies the absence of precise predictive biomarkers and poor understanding of the mechanisms behind tumor resistance as the main obstacles facing ICIs immunotherapy. In the present text, we discuss the dual role of PD-L1 as a biomarker for response to immunotherapy and as an immune checkpoint. The contribution of mass spectrometry-based analysis, particularly the impact of protein post-translational modifications on the performance of this protein is underlined.
Collapse
Affiliation(s)
| | - Pietro Traldi
- Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35100 Padova, Italy; (M.A.); (M.H.)
| | | |
Collapse
|
99
|
Wang T, He M, Guan W. Pyrotinib monotherapy for advanced HER2-positive esophageal adenocarcinoma with trastuzumab resistance and chemotherapy intolerance: a case report and literature review. Discov Oncol 2025; 16:335. [PMID: 40095240 PMCID: PMC11914708 DOI: 10.1007/s12672-025-02049-6] [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: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
HER2-positive advanced esophageal adenocarcinoma (EAC) cases demonstrate a poor prognosis because of drug resistance that develops after standard first-line trastuzumab therapy. The patient was initially diagnosed with stage cT2N1M0 III EAC. He underwent neoadjuvant chemotherapy, radical esophageal resection, and postoperative adjuvant radiotherapy. However, four months after treatment, the lesion relapsed and progressed to the right back, rendering the case inoperable. Pathological analysis revealed HER2 amplification. Given a poor tolerance to chemotherapy, the patient was administered cadonilimab and trastuzumab for three months. Subsequently, the second-line therapy was switched to pyrotinib monotherapy as a salvage treatment. Remarkably, after one month of treatment, the tumor showed significant reduction, with mild toxic side effects. Pyrotinib can be used for salvage later-line therapy in HER2-positive advanced EAC with trastuzumab resistance or poor chemotherapy tolerance, which deserves further promotion.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Mingyuan He
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Wei Guan
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| |
Collapse
|
100
|
Long B, Zhou H, Yu Z, Zhu J, Yang H, Huang Z, Wei D, Chen S, Yang X, Zhao X, Zhang W, Yan H, Guan X, Li L, Zhang G, Yu H, Che S, Gao Z, Jiang X, Luo C, Mao J, Zhao D, Li Y, Jiang Z, Jiao Z. Neoadjuvant cadonilimab plus FLOT chemotherapy in locally advanced gastric/gastroesophageal junction adenocarcinoma: A multicenter, phase 2 study. MED 2025; 6:100531. [PMID: 39536755 DOI: 10.1016/j.medj.2024.10.008] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Treatment with cadonilimab and chemotherapy has shown promise as a first-line treatment for gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, its application in neoadjuvant settings has not yet been documented. METHODS This multicenter, phase 2 trial (ChiCTR2200066893) was conducted at four hospitals across China. Treatment-naive patients with locally advanced G/GEJ adenocarcinoma (cT3/4, N+, M0) and who were human epidermal growth factor receptor 2 negative received 3-cycle or 4-cycle neoadjuvant treatment of cadonilimab plus FLOT (5-fluorouracil, leucovorin, oxaliplatin, and docetaxel) chemotherapy, followed by gastrectomy and 4-cycle adjuvant FLOT chemotherapy. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included major pathological response (MPR), overall response rate (ORR), disease control rate (DCR), R0 resection rate, downstaging rate, and safety. FINDINGS Between December 23, 2022, and December 15, 2023, 32 of 38 patients completed the scheduled treatment, achieving an R0 resection rate of 100% (32/32). The pCR rate was 21.1% (8/38, 90% confidence interval [CI]: 9.7-32.4), and the MPR rate was 44.7% (17/38, 90% CI: 30.9-58.5). Radiological evaluations were available for 28 of 38 patients by blinded independent central review. The ORR was 60.7% (17/28, 90% CI: 44.7-76.7), and the DCR was 100.0% (28/28, 90% CI: 100.0-100.0). Tumor downstaging occurred in 71.9% of patients (23/32), with consistent efficacy across all populations observed in the subgroup analysis. Grade 3 adverse events occurred in 31.6% of patients without severe safety issues. CONCLUSIONS Neoadjuvant cadonilimab plus FLOT chemotherapy treatment exhibits promising efficacy with manageable toxicities in locally advanced G/GEJ adenocarcinoma, providing preliminary evidence for further investigation. FUNDING This study was funded by Akeso Biopharma.
Collapse
Affiliation(s)
- Bo Long
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Huinian Zhou
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeyuan Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Junmin Zhu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hanteng Yang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeping Huang
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Dengwen Wei
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Shigong Chen
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaojun Yang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaoning Zhao
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Wenjuan Zhang
- Lanzhou University Second Hospital, The Radiology Department, Lanzhou, China
| | - Hong Yan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Xiaoying Guan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Long Li
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Gengyuan Zhang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hongwei Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Shengfu Che
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zhongti Gao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiangyan Jiang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Changjiang Luo
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Jie Mao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Da Zhao
- The First Hospital of Lanzhou University, The Oncology Department, Lanzhou, China
| | - Yumin Li
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Zebin Jiang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Zuoyi Jiao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|