301
|
Tittarelli A, Pereda C, Gleisner MA, López MN, Flores I, Tempio F, Lladser A, Achour A, González FE, Durán-Aniotz C, Miranda JP, Larrondo M, Salazar-Onfray F. Long-Term Survival and Immune Response Dynamics in Melanoma Patients Undergoing TAPCells-Based Vaccination Therapy. Vaccines (Basel) 2024; 12:357. [PMID: 38675738 PMCID: PMC11053591 DOI: 10.3390/vaccines12040357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer vaccines present a promising avenue for treating immune checkpoint blockers (ICBs)-refractory patients, fostering immune responses to modulate the tumor microenvironment. We revisit a phase I/II trial using Tumor Antigen-Presenting Cells (TAPCells) (NCT06152367), an autologous antigen-presenting cell vaccine loaded with heat-shocked allogeneic melanoma cell lysates. Initial findings showcased TAPCells inducing lysate-specific delayed-type hypersensitivity (DTH) reactions, correlating with prolonged survival. Here, we extend our analysis over 15 years, categorizing patients into short-term (<36 months) and long-term (≥36 months) survivors, exploring novel associations between clinical outcomes and demographic, genetic, and immunologic parameters. Notably, DTHpos patients exhibit a 53.1% three-year survival compared to 16.1% in DTHneg patients. Extended remissions are observed in long-term survivors, particularly DTHpos/M1cneg patients. Younger age, stage III disease, and moderate immune events also benefit short-term survivors. Immunomarkers like increased C-type lectin domain family 2 member D on CD4+ T cells and elevated interleukin-17A were detected in long-term survivors. In contrast, toll-like receptor-4 D229G polymorphism and reduced CD32 on B cells are associated with reduced survival. TAPCells achieved stable long remissions in 35.2% of patients, especially M1cneg/DTHpos cases. Conclusions: Our study underscores the potential of vaccine-induced immune responses in melanoma, emphasizing the identification of emerging biological markers and clinical parameters for predicting long-term remission.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile;
| | - Cristian Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - María A. Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Mercedes N. López
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Fabián Tempio
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580702, Chile;
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 8580702, Chile
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Fermín E. González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile;
| | - Claudia Durán-Aniotz
- Latin American Brain Health Institute (BrainLat), Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibañez, Santiago 7941169, Chile;
| | | | - Milton Larrondo
- Banco de Sangre, Hospital Clínico de la Universidad de Chile, Santiago 8380453, Chile;
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (C.P.); (M.A.G.); (M.N.L.); (I.F.); (F.T.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden;
- Division of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
302
|
Germer G, Schwartze L, García-Miller J, Balansin-Rigon R, Groth LJ, Rühl I, Patoka P, Zoschke C, Rühl E. Label-free mapping of cetuximab in multi-layered tumor oral mucosa models by atomic force-microscopy-based infrared spectroscopy. Analyst 2024; 149:2122-2130. [PMID: 38436119 DOI: 10.1039/d3an01877f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Sensitive mapping of drugs and drug delivery systems is pivotal for the understanding and improvement of treatment options. Since labeling alters the physicochemical and potentially the pharmacological properties of the molecule of interest, its label-free detection by photothermal expansion is investigated. We report on a proof-of-concept study to map the cetuximab distribution by atomic-force microscopy-based infrared spectroscopy (AFM-IR). The monoclonal antibody cetuximab was applied to a human tumor oral mucosa model, consisting of a tumor epithelium on a lamina propria equivalent. Hyperspectral imaging in the wavenumber regime between 903 cm-1 and 1312 cm-1 and a probing distance between the data points down to 10 × 10 nm are used for determining the local drug distribution. The local distinction of cetuximab from the tissue background is gained by linear combination modeling making use of reference spectra of the drug and untreated models. The results from this approach are compared to principal component analyses, yielding comparable results. Even single molecule detection appears feasible. The results indicate that cetuximab penetrates the cytosol of tumor cells but does not bind to structures in the cell membrane. In conclusion, AFM-IR mapping of cetuximab proved to sensitively determine drug concentrations at an unprecedented spatial resolution without the need for drug labeling.
Collapse
Affiliation(s)
- Gregor Germer
- Physical Chemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany. ruehl@
| | - Leonie Schwartze
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195 Berlin, Germany.
| | - Jill García-Miller
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195 Berlin, Germany.
| | - Roberta Balansin-Rigon
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195 Berlin, Germany.
- School of Pharmaceutical Sciences, University of Campinas, R. Candido Portinari, 200 - Cidade Universitária, Campinas, SP, 13083-871, Brazil
| | - Lucie J Groth
- Physical Chemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany. ruehl@
| | - Isabel Rühl
- Physical Chemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany. ruehl@
| | - Piotr Patoka
- Physical Chemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany. ruehl@
| | - Christian Zoschke
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195 Berlin, Germany.
- Department of Veterinary Medicines, Federal Office of Consumer Protection and Food Safety, Gerichtstr. 49, 13347 Berlin, Germany
| | - Eckart Rühl
- Physical Chemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany. ruehl@
| |
Collapse
|
303
|
Spreafico A, Couselo EM, Irmisch A, Bessa J, Au-Yeung G, Bechter O, Svane IM, Sanmamed MF, Gambardella V, McKean M, Callahan M, Dummer R, Klein C, Umaña P, Justies N, Heil F, Fahrni L, Opolka-Hoffmann E, Waldhauer I, Bleul C, Staack RF, Karanikas V, Fowler S. Phase 1, first-in-human study of TYRP1-TCB (RO7293583), a novel TYRP1-targeting CD3 T-cell engager, in metastatic melanoma: active drug monitoring to assess the impact of immune response on drug exposure. Front Oncol 2024; 14:1346502. [PMID: 38577337 PMCID: PMC10991832 DOI: 10.3389/fonc.2024.1346502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Although checkpoint inhibitors (CPIs) have improved outcomes for patients with metastatic melanoma, those progressing on CPIs have limited therapeutic options. To address this unmet need and overcome CPI resistance mechanisms, novel immunotherapies, such as T-cell engaging agents, are being developed. The use of these agents has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs), which is challenging to predict preclinically and can lead to neutralization of the drug and loss of efficacy. Methods TYRP1-TCB (RO7293583; RG6232) is a T-cell engaging bispecific (TCB) antibody that targets tyrosinase-related protein 1 (TYRP1), which is expressed in many melanomas, thereby directing T cells to kill TYRP1-expressing tumor cells. Preclinical studies show TYRP1-TCB to have potent anti-tumor activity. This first-in-human (FIH) phase 1 dose-escalation study characterized the safety, tolerability, maximum tolerated dose/optimal biological dose, and pharmacokinetics (PK) of TYRP1-TCB in patients with metastatic melanoma (NCT04551352). Results Twenty participants with cutaneous, uveal, or mucosal TYRP1-positive melanoma received TYRP1-TCB in escalating doses (0.045 to 0.4 mg). All participants experienced ≥1 treatment-related adverse event (TRAE); two participants experienced grade 3 TRAEs. The most common toxicities were grade 1-2 cytokine release syndrome (CRS) and rash. Fractionated dosing mitigated CRS and was associated with lower levels of interleukin-6 and tumor necrosis factor-alpha. Measurement of active drug (dual TYPR1- and CD3-binding) PK rapidly identified loss of active drug exposure in all participants treated with 0.4 mg in a flat dosing schedule for ≥3 cycles. Loss of exposure was associated with development of ADAs towards both the TYRP1 and CD3 domains. A total drug PK assay, measuring free and ADA-bound forms, demonstrated that TYRP1-TCB-ADA immune complexes were present in participant samples, but showed no drug activity in vitro. Discussion This study provides important insights into how the use of active drug PK assays, coupled with mechanistic follow-up, can inform and enable ongoing benefit/risk assessment for individuals participating in FIH dose-escalation trials. Translational studies that lead to a better understanding of the underlying biology of cognate T- and B-cell interactions, ultimately resulting in ADA development to novel biotherapeutics, are needed.
Collapse
Affiliation(s)
- Anna Spreafico
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Eva Muñoz Couselo
- Department of Medical Oncology, Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Anja Irmisch
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Juliana Bessa
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - George Au-Yeung
- Department of Medical Oncology, Peter MacCallum Cancer Center and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Oliver Bechter
- Department of General Medical Oncology, Universitair Ziekenhuis (UZ), Leuven, Leuven, Belgium
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy and Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Miguel F. Sanmamed
- Department of Medical Oncology, Clínica Universidad de Navarra and Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Valentina Gambardella
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Meredith McKean
- Sarah Cannon Research Institute at Tennessee Oncology, Nashville, TN, United States
| | - Margaret Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Pablo Umaña
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Nicole Justies
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Florian Heil
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Linda Fahrni
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Eugenia Opolka-Hoffmann
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Inja Waldhauer
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Conrad Bleul
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Roland F. Staack
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Vaios Karanikas
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Stephen Fowler
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
304
|
Meléndez-Vázquez NM, Nguyen TT, Fan X, López-Rivas AR, Fueyo J, Gomez-Manzano C, Godoy-Vitorino F. Gut microbiota composition is associated with the efficacy of Delta-24-RGDOX in malignant gliomas. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200787. [PMID: 38596290 PMCID: PMC10951704 DOI: 10.1016/j.omton.2024.200787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Glioblastoma, the most common primary brain tumor, has a 6.8% survival rate 5 years post diagnosis. Our team developed an oncolytic adenovirus with an OX-40L expression cassette named Delta-24-RGDOX. While studies have revealed the interaction between the gut microbiota and immunotherapy agents, there are no studies linking the gut microbiota with viroimmunotherapy efficacy. We hypothesize that gut bacterial signatures will be associated with oncolytic viral therapy efficacy. To test this hypothesis, we evaluated the changes in gut microbiota in two mouse cohorts: (1) GSC-005 glioblastoma-bearing mice treated orally with indoximod, an immunotherapeutic agent, or with Delta-24-RGDOX by intratumoral injection and (2) a mouse cohort harboring GL261-5 tumors used to mechanistically evaluate the importance of CD4+ T cells in relation to viroimmunotherapy efficacy. Microbiota assessment indicated significant differences in the structure of the gut bacterial communities in viroimmunotherapy-treated animals with higher survival compared with control or indoximod-treated animals. Moreover, viroimmunotherapy-treated mice with prolonged survival had a higher abundance of Bifidobacterium. The CD4+ T cell depletion was associated with gut dysbiosis, lower mouse survival, and lower antitumor efficacy of the therapy. These findings suggest that microbiota modulation along the gut-glioma axis contributes to the clinical efficacy and patient survival of viroimmunotherapy treated animals.
Collapse
Affiliation(s)
- Natalie M. Meléndez-Vázquez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan 00918 PR, USA
| | - Teresa T. Nguyen
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrés R. López-Rivas
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan 00918 PR, USA
| |
Collapse
|
305
|
Chowaniec H, Ślubowska A, Mroczek M, Borowczyk M, Braszka M, Dworacki G, Dobosz P, Wichtowski M. New hopes for the breast cancer treatment: perspectives on the oncolytic virus therapy. Front Immunol 2024; 15:1375433. [PMID: 38576614 PMCID: PMC10991781 DOI: 10.3389/fimmu.2024.1375433] [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: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
Oncolytic virus (OV) therapy has emerged as a promising frontier in cancer treatment, especially for solid tumours. While immunotherapies like immune checkpoint inhibitors and CAR-T cells have demonstrated impressive results, their limitations in inducing complete tumour regression have spurred researchers to explore new approaches targeting tumours resistant to current immunotherapies. OVs, both natural and genetically engineered, selectively replicate within cancer cells, inducing their lysis while sparing normal tissues. Recent advancements in clinical research and genetic engineering have enabled the development of targeted viruses that modify the tumour microenvironment, triggering anti-tumour immune responses and exhibiting synergistic effects with other cancer therapies. Several OVs have been studied for breast cancer treatment, including adenovirus, protoparvovirus, vaccinia virus, reovirus, and herpes simplex virus type I (HSV-1). These viruses have been modified or engineered to enhance their tumour-selective replication, reduce toxicity, and improve oncolytic properties.Newer generations of OVs, such as Oncoviron and Delta-24-RGD adenovirus, exhibit heightened replication selectivity and enhanced anticancer effects, particularly in breast cancer models. Clinical trials have explored the efficacy and safety of various OVs in treating different cancers, including melanoma, nasopharyngeal carcinoma, head and neck cancer, and gynecologic malignancies. Notably, Talimogene laherparepvec (T-VEC) and Oncorine have. been approved for advanced melanoma and nasopharyngeal carcinoma, respectively. However, adverse effects have been reported in some cases, including flu-like symptoms and rare instances of severe complications such as fistula formation. Although no OV has been approved specifically for breast cancer treatment, ongoing preclinical clinical trials focus on four groups of viruses. While mild adverse effects like low-grade fever and nausea have been observed, the effectiveness of OV monotherapy in breast cancer remains insufficient. Combination strategies integrating OVs with chemotherapy, radiotherapy, or immunotherapy, show promise in improving therapeutic outcomes. Oncolytic virus therapy holds substantial potential in breast cancer treatment, demonstrating safety in trials. Multi-approach strategies combining OVs with conventional therapies exhibit more promising therapeutic effects than monotherapy, signalling a hopeful future for OV-based breast cancer treatments.
Collapse
Affiliation(s)
- Hanna Chowaniec
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Antonina Ślubowska
- Department of Biostatistics and Research Methodology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University of Warsaw, Warsaw, Poland
| | - Magdalena Mroczek
- Department of Neurology, University Hospital Basel, Univeristy of Basel, Basel, Switzerland
| | - Martyna Borowczyk
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Małgorzata Braszka
- Faculty of Medical Sciences, University College London Medical School, London, United Kingdom
| | - Grzegorz Dworacki
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Chair of Patomorphology and Clinical Immunology, Poznań University of Medical Sciences, Poznan, Poland
| | - Paula Dobosz
- University Centre of Cancer Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Mateusz Wichtowski
- Surgical Oncology Clinic, Institute of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
306
|
Jiang Y, Zheng Y, Zhang YW, Kong S, Dong J, Wang F, Ziman B, Gery S, Hao JJ, Zhou D, Zhou J, Ho AS, Sinha UK, Chen J, Zhang S, Yin C, Wei DD, Hazawa M, Pan H, Lu Z, Wei WQ, Wang MR, Koeffler HP, Lin DC, Jiang YY. Reciprocal inhibition between TP63 and STAT1 regulates anti-tumor immune response through interferon-γ signaling in squamous cancer. Nat Commun 2024; 15:2484. [PMID: 38509096 PMCID: PMC10954759 DOI: 10.1038/s41467-024-46785-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Squamous cell carcinomas (SCCs) are common and aggressive malignancies. Immune check point blockade (ICB) therapy using PD-1/PD-L1 antibodies has been approved in several types of advanced SCCs. However, low response rate and treatment resistance are common. Improving the efficacy of ICB therapy requires better understanding of the mechanism of immune evasion. Here, we identify that the SCC-master transcription factor TP63 suppresses interferon-γ (IFNγ) signaling. TP63 inhibition leads to increased CD8+ T cell infiltration and heighten tumor killing in in vivo syngeneic mouse model and ex vivo co-culture system, respectively. Moreover, expression of TP63 is negatively correlated with CD8+ T cell infiltration and activation in patients with SCC. Silencing of TP63 enhances the anti-tumor efficacy of PD-1 blockade by promoting CD8+ T cell infiltration and functionality. Mechanistically, TP63 and STAT1 mutually suppress each other to regulate the IFNγ signaling by co-occupying and co-regulating their own promoters and enhancers. Together, our findings elucidate a tumor-extrinsic function of TP63 in promoting immune evasion of SCC cells. Over-expression of TP63 may serve as a biomarker predicting the outcome of SCC patients treated with ICB therapy, and targeting TP63/STAT/IFNγ axis may enhance the efficacy of ICB therapy for this deadly cancer.
Collapse
Affiliation(s)
- Yuan Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Yueyuan Zheng
- Clinical Big Data Research Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuan-Wei Zhang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Shuai Kong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jinxiu Dong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Fei Wang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Benjamin Ziman
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sigal Gery
- Department of Medicine, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jia-Jie Hao
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dan Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Institutes of Physical Science and Technology, Anhui University, Hefei, 230601, China
| | - Jianian Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Allen S Ho
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Uttam K Sinha
- Department of otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jian Chen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Shuo Zhang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Chuntong Yin
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Dan-Dan Wei
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Masaharu Hazawa
- Cell-Bionomics Research Unit, Innovative Integrated Bio-Research Core, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Huaguang Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Wen-Qiang Wei
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - H Phillip Koeffler
- Department of Medicine, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - De-Chen Lin
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Yan-Yi Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
307
|
Scheuermann S, Kristmann B, Engelmann F, Nuernbergk A, Scheuermann D, Koloseus M, Abed T, Solass W, Seitz CM. Unveiling spatial complexity in solid tumor immune microenvironments through multiplexed imaging. Front Immunol 2024; 15:1383932. [PMID: 38566984 PMCID: PMC10985204 DOI: 10.3389/fimmu.2024.1383932] [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: 02/08/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Deciphering cellular components and the spatial interaction network of the tumor immune microenvironment (TIME) of solid tumors is pivotal for understanding biologically relevant cross-talks and, ultimately, advancing therapies. Multiplexed tissue imaging provides a powerful tool to elucidate spatial complexity in a holistic manner. We established and cross-validated a comprehensive immunophenotyping panel comprising over 121 markers for multiplexed tissue imaging using MACSima™ imaging cyclic staining (MICS) alongside an end-to-end analysis workflow. Applying this panel and workflow to primary cancer tissues, we characterized tumor heterogeneity, investigated potential therapeutical targets, conducted in-depth profiling of cell types and states, sub-phenotyped T cells within the TIME, and scrutinized cellular neighborhoods of diverse T cell subsets. Our findings highlight the advantage of spatial profiling, revealing immunosuppressive molecular signatures of tumor-associated myeloid cells interacting with neighboring exhausted, PD1high T cells in the TIME of hepatocellular carcinoma (HCC). This study establishes a robust framework for spatial exploration of TIMEs in solid tumors and underscores the potency of multiplexed tissue imaging and ultra-deep cell phenotyping in unraveling clinically relevant tumor components.
Collapse
Affiliation(s)
- Sophia Scheuermann
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), partner site Tuebingen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Tuebingen, Tuebingen, Germany
| | - Beate Kristmann
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
| | - Fabienne Engelmann
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
| | - Alice Nuernbergk
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
| | - David Scheuermann
- School of Business and Economics, Faculty of Economics and Social Sciences, University of Tuebingen, Tuebingen, Germany
| | - Marie Koloseus
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
| | - Tayeb Abed
- Institute of Pathology and Neuropathology, University Hospital Tuebingen and Comprehensive Cancer Center, Tuebingen, Germany
| | - Wiebke Solass
- Institute of Tissue Medicine and Pathology (ITMP), University of Bern, Bern, Switzerland
| | - Christian M. Seitz
- Department of Haematology, Oncology, Gastroenterology, Nephrology, Rheumatology, University Children’s Hospital Tuebingen, Tuebingen, Germany
- iFIT Cluster of Excellence EXC 2180 ‘Image-Guided and Functionally Instructed Tumor Therapies’, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), partner site Tuebingen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
308
|
Nejo T, Wang L, Leung KK, Wang A, Lakshmanachetty S, Gallus M, Kwok DW, Hong C, Chen LH, Carrera DA, Zhang MY, Stevers NO, Maldonado GC, Yamamichi A, Watchmaker PB, Naik A, Shai A, Phillips JJ, Chang SM, Wiita AP, Wells JA, Costello JF, Diaz AA, Okada H. Challenges in the discovery of tumor-specific alternative splicing-derived cell-surface antigens in glioma. Sci Rep 2024; 14:6362. [PMID: 38493204 PMCID: PMC10944514 DOI: 10.1038/s41598-024-56684-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/09/2024] [Indexed: 03/18/2024] Open
Abstract
Despite advancements in cancer immunotherapy, solid tumors remain formidable challenges. In glioma, profound inter- and intra-tumoral heterogeneity of antigen landscape hampers therapeutic development. Therefore, it is critical to consider alternative sources to expand the repertoire of targetable (neo-)antigens and improve therapeutic outcomes. Accumulating evidence suggests that tumor-specific alternative splicing (AS) could be an untapped reservoir of antigens. In this study, we investigated tumor-specific AS events in glioma, focusing on those predicted to generate major histocompatibility complex (MHC)-presentation-independent, cell-surface antigens that could be targeted by antibodies and chimeric antigen receptor-T cells. We systematically analyzed bulk RNA-sequencing datasets comparing 429 tumor samples (from The Cancer Genome Atlas) and 9166 normal tissue samples (from the Genotype-Tissue Expression project), and identified 13 AS events in 7 genes predicted to be expressed in more than 10% of the patients, including PTPRZ1 and BCAN, which were corroborated by an external RNA-sequencing dataset. Subsequently, we validated our predictions and elucidated the complexity of the isoforms using full-length transcript amplicon sequencing on patient-derived glioblastoma cells. However, analyses of the RNA-sequencing datasets of spatially mapped and longitudinally collected clinical tumor samples unveiled remarkable spatiotemporal heterogeneity of the candidate AS events. Furthermore, proteomics analysis did not reveal any peptide spectra matching the putative antigens. Our investigation illustrated the diverse characteristics of the tumor-specific AS events and the challenges of antigen exploration due to their notable spatiotemporal heterogeneity and elusive nature at the protein levels. Redirecting future efforts toward intracellular, MHC-presented antigens could offer a more viable avenue.
Collapse
Affiliation(s)
- Takahide Nejo
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Lin Wang
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Senthilnath Lakshmanachetty
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Marco Gallus
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Darwin W Kwok
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Lee H Chen
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Diego A Carrera
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Michael Y Zhang
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Nicholas O Stevers
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Gabriella C Maldonado
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Akane Yamamichi
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Payal B Watchmaker
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Akul Naik
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Anny Shai
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Aaron A Diaz
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco (UCSF), 1450 3Rd Street, Box 0520, San Francisco, CA, 94158, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
309
|
Zhu R, Chen TZ, Sun MT, Zhu CR. Advanced cervix cancer patient with chemotherapy-induced grade IV myelosuppression achieved complete remission with cadonilimab: A case report. World J Clin Cases 2024; 12:1510-1516. [PMID: 38576806 PMCID: PMC10989456 DOI: 10.12998/wjcc.v12.i8.1510] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/18/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The prognosis for patients with advanced metastatic cervix cancer (MCC) is poor, and this disease continues to pose a considerable therapeutic challenge. Despite the administration of first-line regimens consisting of cisplatin, paclitaxel, and bevacizumab, survival rates for patients with metastasis remain poor. The emergence of bispecific antibodies (BsAbs) offers a novel treatment option for patients diagnosed with MCC. CASE SUMMARY In this report, we present a patient with MCC who was treated with cadonilimab monotherapy at a dose of 6 mg/kg every two weeks after chemotherapy was proven to be intolerable. The patient exhibited a sustained complete response for a duration of 6 months, demonstrating an optimistic outlook. CONCLUSION This case illustrates the considerable efficacy of cadonilimab for treating advanced MCC. Therefore, BsAb therapy is a promising strategy for effectively treating patients with advanced MCC and should be considered as an option when patients are intolerant to standard chemotherapy.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Tian-Ze Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Meng-Ting Sun
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Chun-Rong Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| |
Collapse
|
310
|
Liu G, Chen T, Zhang X, Hu B, Shi H. Immune checkpoint inhibitor-associated cardiovascular toxicities: A review. Heliyon 2024; 10:e25747. [PMID: 38434280 PMCID: PMC10907684 DOI: 10.1016/j.heliyon.2024.e25747] [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: 07/19/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionary effects on therapeutic strategies for multiple malignancies. Their efficacy depends on their ability to reactivate the host immune system to fight cancer cells. However, adverse reactions to ICIs are common and involve several organs, limiting their use in clinical practice. Although the incidence of cardiovascular toxicity is relatively low, it is associated with serious consequences and high mortality rates. The primary cardiovascular toxicities include myocarditis, pericarditis, Takotsubo syndrome, arrhythmia, vasculitis, acute coronary syndrome, and venous thromboembolism. Currently, the mechanism underlying ICI-associated cardiovascular toxicity remains unclear and underexplored. The diagnosis and monitoring of ICI-associated cardiovascular toxicities mainly include the following indicators: symptoms, signs, laboratory examination, electrocardiography, imaging, and pathology. Treatments are based on the grade of cardiovascular toxicity and mainly include drug withdrawal, corticosteroid therapy, immunosuppressants, and conventional cardiac treatment. This review focuses on the incidence, underlying mechanisms, clinical manifestations, diagnoses, and treatment strategies.
Collapse
Affiliation(s)
- Guihong Liu
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Chen
- Tao Chen Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Binbin Hu
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huashan Shi
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
311
|
Tran SD, Lin J, Galvez C, Rasmussen LV, Pacheco J, Perottino GM, Rahbari KJ, Miller CD, John JD, Theros J, Vogel K, Dinh PV, Malik S, Ramzan U, Tegtmeyer K, Mohindra N, Johnson JL, Luo Y, Kho A, Sosman J, Walunas TL. Rapid identification of inflammatory arthritis and associated adverse events following immune checkpoint therapy: a machine learning approach. Front Immunol 2024; 15:1331959. [PMID: 38558818 PMCID: PMC10978703 DOI: 10.3389/fimmu.2024.1331959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Immune checkpoint inhibitor-induced inflammatory arthritis (ICI-IA) poses a major clinical challenge to ICI therapy for cancer, with 13% of cases halting ICI therapy and ICI-IA being difficult to identify for timely referral to a rheumatologist. The objective of this study was to rapidly identify ICI-IA patients in clinical data and assess associated immune-related adverse events (irAEs) and risk factors. Methods We conducted a retrospective study of the electronic health records (EHRs) of 89 patients who developed ICI-IA out of 2451 cancer patients who received ICI therapy at Northwestern University between March 2011 to January 2021. Logistic regression and random forest machine learning models were trained on all EHR diagnoses, labs, medications, and procedures to identify ICI-IA patients and EHR codes indicating ICI-IA. Multivariate logistic regression was then used to test associations between ICI-IA and cancer type, ICI regimen, and comorbid irAEs. Results Logistic regression and random forest models identified ICI-IA patients with accuracies of 0.79 and 0.80, respectively. Key EHR features from the random forest model included ICI-IA relevant features (joint pain, steroid prescription, rheumatoid factor tests) and features suggesting comorbid irAEs (thyroid function tests, pruritus, triamcinolone prescription). Compared to 871 adjudicated ICI patients who did not develop arthritis, ICI-IA patients had higher odds of developing cutaneous (odds ratio [OR]=2.66; 95% Confidence Interval [CI] 1.63-4.35), endocrine (OR=2.09; 95% CI 1.15-3.80), or gastrointestinal (OR=2.88; 95% CI 1.76-4.72) irAEs adjusting for demographics, cancer type, and ICI regimen. Melanoma (OR=1.99; 95% CI 1.08-3.65) and renal cell carcinoma (OR=2.03; 95% CI 1.06-3.84) patients were more likely to develop ICI-IA compared to lung cancer patients. Patients on nivolumab+ipilimumab were more likely to develop ICI-IA compared to patients on pembrolizumab (OR=1.86; 95% CI 1.01-3.43). Discussion Our machine learning models rapidly identified patients with ICI-IA in EHR data and elucidated clinical features indicative of comorbid irAEs. Patients with ICI-IA were significantly more likely to also develop cutaneous, endocrine, and gastrointestinal irAEs during their clinical course compared to ICI therapy patients without ICI-IA.
Collapse
Affiliation(s)
- Steven D. Tran
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jean Lin
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Carlos Galvez
- Hematology and Oncology, University of Illinois Health, Chicago, IL, United States
| | - Luke V. Rasmussen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jennifer Pacheco
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | | | - Kian J. Rahbari
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Charles D. Miller
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jordan D. John
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jonathan Theros
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kelly Vogel
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Patrick V. Dinh
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sara Malik
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Umar Ramzan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kyle Tegtmeyer
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Nisha Mohindra
- Department of Medicine, Division of Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| | - Jodi L. Johnson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
- Departments of Pathology and Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Abel Kho
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Medicine, Division of General Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeffrey Sosman
- Department of Medicine, Division of Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| | - Theresa L. Walunas
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Medicine, Division of General Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
312
|
Beasley GM, Terando AM. Articles from 2022 to 2023 to Inform Your Cancer Practice: Melanoma. Ann Surg Oncol 2024; 31:1851-1856. [PMID: 38071709 DOI: 10.1245/s10434-023-14702-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 02/08/2024]
Abstract
Modern effective systemic therapy for melanoma includes two important classes of treatment: immune checkpoint inhibitors (ICIs), comprising inhibitors of cytotoxic T-lymphocyte antigen 4, programmed cell death receptor 1, and lymphocyte-activation gene 3; and small molecule BRAF/MEK inhibitor therapy. These treatments have revolutionized the management of patients with advanced melanoma and have dramatically improved clinical outcomes. The melanoma treatment landscape continues to evolve as outcome data from completed trials continue to mature and as newer studies begin to report data. In 2022 and 2023, longer-term follow-up data for established single-agent ICI therapy has been published improving our understanding of both efficacy and durability of treatment responses. A trial of a novel combination ICI therapy has demonstrated enhanced efficacy, and a study examining the order/sequence of ICI therapy versus BRAF/MEK inhibitor therapy for first-line treatment of metastatic melanoma showed that survival is improved when patients start with ICI therapy. As the indications for these therapies have expanded to the adjuvant and neoadjuvant space, we also saw the publication of 5-year results of adjuvant therapy in resected stage III patients, new data on the role of adjuvant therapy in resected stage IIB and IIC patients, and, finally, a practice-changing trial demonstrating improved outcomes using a neoadjuvant approach for patients with macroscopic disease amenable to surgical resection. In this article, we review these articles and highlight key elements for surgical oncologists.
Collapse
Affiliation(s)
- Georgia M Beasley
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Alicia M Terando
- Department of Surgery, Cedars-Sinai Health System, Los Angeles, CA, USA
| |
Collapse
|
313
|
Shah V, Panchal V, Shah A, Vyas B, Agrawal S, Bharadwaj S. Immune checkpoint inhibitors in metastatic melanoma therapy (Review). MEDICINE INTERNATIONAL 2024; 4:13. [PMID: 38410760 PMCID: PMC10895472 DOI: 10.3892/mi.2024.137] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024]
Abstract
An increase in the incidence of melanoma has been observed in recent decades, which poses a significant challenge due to its poor prognosis in the advanced and metastatic stages. Previously, chemotherapy and high doses of interleukin-2 were available treatments for melanoma; however, they offered limited survival benefits and were associated with severe toxicities. The treatment of metastatic melanoma has been transformed by new developments in immunotherapy. Immune checkpoint inhibitors (ICIs), monoclonal antibodies that target cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and its ligand, PDL-1, have emerged as promising therapeutic options. Commonly used ICIs, such as ipilimumab, nivolumab and pembrolizumab, have been found to be associated with an improved median overall survival, recurrence-free survival and response rates compared to traditional chemotherapies. Combination therapies involving different types of ICIs, such as anti-PD1 with anti-CTLA-4, have further enhanced the overall survival and response rates by targeting various phases of T-cell activation. Additionally, the development of novel biomarkers has facilitated the assessment of responses to ICI therapy, with tissue and serum-based prognostic and predictive biomarkers now available. The increased response observed with ICIs also provides potential for immune-related adverse effects on various organ systems. Further research is required to evaluate the efficacy and safety of various combinations of ICIs, while ongoing clinical trials explore the potential of newer ICIs. Concerns regarding the development of resistance to ICIs also warrant attention. The present review summarizes and discusses the advent of ICIs with a marked significant breakthrough in the treatment of metastatic melanoma, providing improved outcomes compared to traditional therapies.
Collapse
Affiliation(s)
- Vedant Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Viraj Panchal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Abhi Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Bhavya Vyas
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Siddharth Agrawal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Sanket Bharadwaj
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| |
Collapse
|
314
|
Chick RC, Ruff SM, Pawlik TM. Neoadjuvant systemic therapy for hepatocellular carcinoma. Front Immunol 2024; 15:1355812. [PMID: 38495884 PMCID: PMC10940409 DOI: 10.3389/fimmu.2024.1355812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Surgical resection and liver transplant remain the only curative therapies for most patients with hepatocellular carcinoma (HCC). Systemic therapy options have typically been ineffective, but recent advances, such as the combination of immune checkpoint inhibitors and targeted therapies, have shown great promise. Neoadjuvant systemic therapy in resectable or locally advanced HCC is under active investigation with encouraging results in small, early-phase trials. Many of these completed and ongoing trials include combinations of systemic therapy (e.g. immune checkpoint inhibitors, tyrosine kinase inhibitors), transarterial therapies, and radiation. Despite early successes, larger trials with evaluation of long-term oncologic outcomes are needed to determine the role of neoadjuvant systemic therapy in patients with HCC who may be eligible for curative intent surgery or transplant.
Collapse
Affiliation(s)
| | | | - Timothy M. Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
315
|
Ressler JM, Tomasich E, Hatziioannou T, Ringl H, Heller G, Silmbrod R, Gottmann L, Starzer AM, Zila N, Tschandl P, Hoeller C, Preusser M, Berghoff AS. DNA Methylation Signatures Correlate with Response to Immune Checkpoint Inhibitors in Metastatic Melanoma. Target Oncol 2024; 19:263-275. [PMID: 38401029 DOI: 10.1007/s11523-024-01041-4] [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: 02/02/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND DNA methylation profiles have emerged as potential predictors of therapeutic response in various solid tumors. OBJECTIVE This study aimed to analyze the DNA methylation profiles of patients with stage IV metastatic melanoma undergoing first-line immune checkpoint inhibitor treatment and evaluate their correlation with a radiological response according to immune-related Response Evaluation Criteria in Solid Tumors (iRECIST). METHODS A total of 81 tissue samples from 71 patients with metastatic melanoma (27 female, 44 male) were included in this study. We utilized Illumina Methylation EPIC Beadchips to retrieve their genome-wide methylation profile by interrogating >850,000 CpG sites. Clustering based on the 500 most differentially methylated genes was conducted to identify distinct methylation patterns associated with immune checkpoint inhibitor response. Results were further aligned with an independent, previously published data set. RESULTS The median progression-free survival was 8.5 months (range: 0-104.1 months), and the median overall survival was 30.6 months (range: 0-104.1 months). Objective responses were observed in 29 patients (40.8%). DNA methylation profiling revealed specific signatures that correlated with radiological response to immune checkpoint inhibitors. Three distinct clusters were identified based on the methylation patterns of the 500 most differentially methylated genes. Cluster 1 (12/12) and cluster 2 (12/24) exhibited a higher proportion of responders, while cluster 3 (39/45) predominantly consisted of non-responders. In the validation data set, responders also showed more frequent hypomethylation although differences in the data sets limit the interpretation. CONCLUSIONS These findings suggest that DNA methylation profiling of tumor tissues might serve as a predictive biomarker for immune checkpoint inhibitor response in patients with metastatic melanoma. Further validation studies are warranted to confirm the efficiency of DNA methylation profiling as a predictive tool in the context of immunotherapy for metastatic melanoma.
Collapse
Affiliation(s)
| | - Erwin Tomasich
- Department of Medicine I, Division of Oncology, Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Teresa Hatziioannou
- Department of Medicine I, Division of Oncology, Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Helmut Ringl
- Wiener Gesundheitsverbund, Klinik Donaustadt, Vienna, Austria
| | - Gerwin Heller
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Rita Silmbrod
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Lynn Gottmann
- Department of Medicine I, Division of Oncology, Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Philipp Tschandl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christoph Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Anna Sophie Berghoff
- Department of Medicine I, Division of Oncology, Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
316
|
Benhima N, Belbaraka R, Langouo Fontsa MD. Single agent vs combination immunotherapy in advanced melanoma: a review of the evidence. Curr Opin Oncol 2024; 36:69-73. [PMID: 38193381 DOI: 10.1097/cco.0000000000001014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is to outline the current landscape of advanced melanoma treatment options, provide insights on selecting combination therapies within different clinical scenarios, capture clinical relevance of anti-programmed cell death protein 1 (PD-1) monotherapy, and explore the unmet needs with immune check-point inhibitors (ICI) in advanced melanoma. RECENT FINDINGS ICI based treatment consisted of single agent ICI or dual combination ICI-ICI is the standard of care of front-line treatment of metastatic or unresectable melanoma. PD-1 inhibitors (Pembrolizumab and Nivolumab) improved progression free survival (PFS) and overall survival (OS) compared to chemotherapy and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) inhibitors (Ipilimumab and Tremelimumab). The dual ICI combination (Nivolumab and Ipilimumab) provided profound and durable responses better than monotherapy, and the longest overall survival ever achieved in advanced disease, including in patients with murine sarcoma viral oncogene homolog B (BRAF)-mutated disease, but at the cost of a high risk of severe toxicity. The new dual blockage of LAG-3 and PD-1 (Nivolumab-Relatlimab) emerges as a valid option with promising efficacy outcomes and a favourable toxicity profile. Mature survival data is still needed to capture the real benefit. SUMMARY These new plethora of options pose new challenges not only for optimal treatment sequencing strategies but especially for management of adverse effects, endorsing the need to integrate a holistic and personalized approach for patient care.
Collapse
Affiliation(s)
- Nada Benhima
- Medical Oncology Department, Mohammed VI University Hospital, Marrakech, Morocco
- Medical Oncology Clinic, Jules Bordet Institute, Brussels, Belgium
| | - Rhizlane Belbaraka
- Medical Oncology Department, Mohammed VI University Hospital, Marrakech, Morocco
| | | |
Collapse
|
317
|
Namikawa K, Nakano E, Ogata D, Yamazaki N. Long-term survival with systemic therapy in the last decade: Can melanoma be cured? J Dermatol 2024; 51:343-352. [PMID: 38358050 PMCID: PMC11484129 DOI: 10.1111/1346-8138.17147] [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: 01/14/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Immune checkpoint inhibitors have been shown to prolong survival of patients with several types of cancer, and the finding was first established in melanoma. Previously, systemic therapy for advanced melanoma aimed only at tumor control and palliation of symptoms. However, in recent years, some patients who received systemic therapy have achieved a complete response and survived without continuous treatment for more than several years. This review discusses the long-term survival rates achieved with currently used systemic therapies and their future perspectives. Long-term survival is currently most likely to be achieved with the use of the standard-dose combination of nivolumab plus ipilimumab, however, this regimen is associated with a high frequency of serious or persistent immune-related adverse events. Several new anti-PD-1-based combination therapies with a better risk-benefit balance are currently under development. Although the acral and mucosal subtypes tend to be less responsive to immune checkpoint inhibitors, anti-PD-1-based combination therapy should continue to be investigated for these subtypes owing to its potential for better long-term survival. With the development of efficacious immunotherapy and targeted therapy, it is important to determine the optimal duration of systemic therapy to avoid unnecessary health and financial burdens as well as to improve efforts to support long-term cancer survivors. As the goal of systemic therapy shifts from tumor control to long-term survival, in future clinical trials, long-term clinical outcomes should be evaluated to assess the benefits of novel agents.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Eiji Nakano
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Dai Ogata
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
318
|
Kim HD, Yeh CY, Chang YC, Kim CH. Dawn era for revisited cancer therapy by innate immune system and immune checkpoint inhibitors. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167019. [PMID: 38211726 DOI: 10.1016/j.bbadis.2024.167019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Immunotherapy is a promising therapeutic strategy for cancer. However, it shows limited efficacy against certain tumor types. The activation of innate immunity can suppress tumors by mitigating inflammatory and malignant behaviors through immune surveillance. The tumor microenvironment, which is composed of immune cells and cancer cells, plays a crucial role in determining the outcomes of immunotherapy. Relying solely on immune checkpoint inhibitors is not an optimal approach. Instead, there is a need to consider the use of a combination of immune checkpoint inhibitors with other modulators of the innate immune system to improve the tumor microenvironment. This can be achieved through methods such as immune cell antigen presentation and recognition. In this review, we delve into the significance of innate immune cells in tumor regression, as well as the role of the interaction of tumor cells with innate immune cells in evading host immune surveillance. These findings pave the way for the next chapter in the field of immunotherapy.
Collapse
Affiliation(s)
- Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Chia-Ying Yeh
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chan Chang
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea; Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea.
| |
Collapse
|
319
|
Gunjur A, Shao Y, Rozday T, Klein O, Mu A, Haak BW, Markman B, Kee D, Carlino MS, Underhill C, Frentzas S, Michael M, Gao B, Palmer J, Cebon J, Behren A, Adams DJ, Lawley TD. A gut microbial signature for combination immune checkpoint blockade across cancer types. Nat Med 2024; 30:797-809. [PMID: 38429524 PMCID: PMC10957475 DOI: 10.1038/s41591-024-02823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/12/2024] [Indexed: 03/03/2024]
Abstract
Immune checkpoint blockade (ICB) targeting programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte protein 4 (CTLA-4) can induce remarkable, yet unpredictable, responses across a variety of cancers. Studies suggest that there is a relationship between a cancer patient's gut microbiota composition and clinical response to ICB; however, defining microbiome-based biomarkers that generalize across cohorts has been challenging. This may relate to previous efforts quantifying microbiota to species (or higher taxonomic rank) abundances, whereas microbial functions are often strain specific. Here, we performed deep shotgun metagenomic sequencing of baseline fecal samples from a unique, richly annotated phase 2 trial cohort of patients with diverse rare cancers treated with combination ICB (n = 106 discovery cohort). We demonstrate that strain-resolved microbial abundances improve machine learning predictions of ICB response and 12-month progression-free survival relative to models built using species-rank quantifications or comprehensive pretreatment clinical factors. Through a meta-analysis of gut metagenomes from a further six comparable studies (n = 364 validation cohort), we found cross-cancer (and cross-country) validity of strain-response signatures, but only when the training and test cohorts used concordant ICB regimens (anti-PD-1 monotherapy or combination anti-PD-1 plus anti-CTLA-4). This suggests that future development of gut microbiome diagnostics or therapeutics should be tailored according to ICB treatment regimen rather than according to cancer type.
Collapse
Affiliation(s)
- Ashray Gunjur
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK.
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK.
| | - Yan Shao
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Timothy Rozday
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Oliver Klein
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
- Department of Medical Oncology, Austin Health, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Andre Mu
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Bastiaan W Haak
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Ben Markman
- Department of Medical Oncology, Monash Health, Melbourne, Victoria, Australia
- Department of Medical Oncology, Alfred Health, Melbourne, Victoria, Australia
- School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Damien Kee
- Department of Medical Oncology, Austin Health, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Rare Cancer Laboratory, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Matteo S Carlino
- Department of Medical Oncology, Blacktown and Westmead Hospitals, Sydney, New South Wales, Australia
- Melanoma Institute of Australia, University of Sydney, Sydney, New South Wales, Australia
| | - Craig Underhill
- Border Medical Oncology and Haematology Research Unit, Albury-Wodonga Regional Cancer Centre, Albury-Wodonga, New South Wales, Australia
- Rural Medical School, University of New South Wales, Albury, New South Wales, Australia
| | - Sophia Frentzas
- Department of Medical Oncology, Monash Health, Melbourne, Victoria, Australia
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Bo Gao
- Department of Medical Oncology, Blacktown and Westmead Hospitals, Sydney, New South Wales, Australia
| | - Jodie Palmer
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
- Department of Medical Oncology, Austin Health, Melbourne, Victoria, Australia
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Melbourne, Victoria, Australia
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
320
|
Abstract
ABSTRACT Traditional chemotherapy has been ineffective in the treatment of metastatic melanoma. Until the use of checkpoint inhibitors, patients had very limited survival. Since the original US Food and Drug Administration approval of ipilimumab over a decade ago, the armamentarium of immunotherapeutic agents has expanded to include programmed cell death protein 1 and lymphocyte activation gene 3 antibodies, requiring a nuanced approach to the selection of frontline treatments, managing patients through recurrence and progression, and determining length of therapy. Herein, we review the existing evidence supporting current standard immunotherapy regimens and discuss the clinical decision-making involved in treating patients with metastatic melanoma with checkpoint inhibitors.
Collapse
Affiliation(s)
- Maya Dimitrova
- From the Laura and Isaac Perlmutter Comprehensive Cancer Center, NYU Grossman School of Medicine, New York, NY
| | | |
Collapse
|
321
|
Staeger R, Martínez-Gómez JM, Turko P, Ramelyte E, Kraehenbuehl L, Del Prete V, Hasan Ali O, Levesque MP, Dummer R, Nägeli MC, Mangana J. Real-World Data on Clinical Outcomes and Treatment Management of Advanced Melanoma Patients: Single-Center Study of a Tertiary Cancer Center in Switzerland. Cancers (Basel) 2024; 16:854. [PMID: 38473216 DOI: 10.3390/cancers16050854] [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: 01/31/2024] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) and BRAF/MEK inhibitors (BRAF/MEKi) have drastically changed the outcomes of advanced melanoma patients in both the resectable/adjuvant and unresectable/metastatic setting. In this follow-up analysis of real-world data, we aimed to investigate the clinical management and outcomes of advanced melanoma patients in a tertiary referral center in Switzerland approximately a decade after the introduction of ICIs and BRAF/MEKi into clinical use. Moreover, we aimed to compare the results with seminal phase 3 trials and to identify areas of high unmet clinical need. METHODS This single-center retrospective cohort study analyzed the melanoma registry of the University Hospital Zurich, a tertiary cancer center in Switzerland, and included patients treated in the resectable/adjuvant (n = 331) or unresectable/metastatic setting (n = 375). RESULTS In the resectable setting, adjuvant anti-PD1 or BRAF/MEKi showed a 3-year relapse-free survival (RFS) of 53% and 67.6%, respectively, and the overall median RFS was 50 months. Patients with lymph node plus in-transit metastases or with distant metastases prior to commencing adjuvant treatment had a significantly reduced overall survival (OS). In 10.9% of patients, the treatment was stopped due to toxicity, which did not affect RFS/OS, unless the duration of the treatment was <3 months. Following a relapse of the disease during the first adjuvant treatment, the median progression-free survival (PFS2) was only 6.6 months; outcomes were particularly poor for relapses that were unresectable (median PFS2 3.9 months) or occurred within the first 2 months (median PFS2 2.7 months). A second adjuvant treatment for patients with resectable relapses still showed efficacy (median RFS2 43.7 months). Elevated LDH levels in patients with an unresectable relapse was correlated with a strong reduction in OS2 (HR 9.84, p = 0.018). In the unresectable setting, first-line anti-PD1, anti-CTLA4/PD1 combination, or BRAF/MEKi showed a 5-year OS of 46.5%, 52.4%, and 49.2%, respectively. In a multivariate analysis, elevated LDH levels or the presence of brain metastases substantially shortened OS (HR > 1.78, p < 0.035). There was a non-significant trend for the improved survival of patients treated with anti-CTLA4/PD1 compared to anti-PD1 (HR 0.64, p = 0.15). After a progression on first-line therapy, the median OS2 was reduced to below two years. Elevated LDH (HR 4.65, p < 0.001) levels and widespread disease with at least three metastatic sites, particularly bone metastases (HR 2.62, p = 0.026), affected OS2. CONCLUSION Our study offers real-world insights into the clinical management, treatment patterns, and outcomes of advanced melanoma patients in both the adjuvant and unresectable setting. Early relapses in patients undergoing adjuvant treatment pose a particular challenge but these patients are generally excluded from first-line trials. The approved first-line metastatic treatments are highly effective in the real-world setting with 5-year OS rates around 50%. However, outcomes remain poor for patients with brain metastases or who fail first-line treatment.
Collapse
Affiliation(s)
- Ramon Staeger
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Julia M Martínez-Gómez
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Patrick Turko
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Egle Ramelyte
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Lukas Kraehenbuehl
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Valerio Del Prete
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Omar Hasan Ali
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mitchell P Levesque
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mirjam C Nägeli
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Joanna Mangana
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
322
|
Isaacs DJ, Kathuria-Prakash N, Hilder R, Lechner MG, Drakaki A. Risk of severe immune-related adverse events in cancer patients with pre-existing autoimmunity receiving immune checkpoint inhibitor therapy. CURRENT CANCER REPORTS 2024; 5:168-180. [PMID: 39397890 PMCID: PMC11469208 DOI: 10.25082/ccr.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Purpose To evaluate the frequency and severity of irAEs in patients with pre-existing autoimmunity, including irAE-related morbidity and mortality, irAE-related management and resolution, and outcome of ICI rechallenge, to better understand the treatment options for this vulnerable patient population. Methods We designed a retrospective, single-center, case-control study at a large, academic medical center to evaluate the incidence and severity of irAEs in patients with pre-existing autoimmunity compared to controls. Controls were matched 2:1 for age, sex, cancer histology, and ICI class. Patients were identified with ICD 9 and 10 codes followed by manual chart extraction. Cases were defined as patients with pre-existing, systemic autoimmunity. The primary outcome was severe irAE (Grade 3 or higher by Common Terminology Criteria for Adverse Events) within 6 months of ICI therapy. Secondary outcomes included response to ICIs, resolution of the irAE, ICI rechallenge success, and survival. Statistical analyses were performed by Chi-square, Fishers exact, Mann-Whitney, and Log-rank tests. Results Of 3,130 patients treated with ICIs from 2015-2021, 28 cases with pre-existing autoimmune disease were identified and were matched with 56 controls. Pre-existing autoimmune conditions included antiphospholipid syndrome, inflammatory polyarthritis, juvenile rheumatoid arthritis, multiple sclerosis, psoriatic arthritis, rheumatoid arthritis, and type I diabetes. Multiple cancer histologies, including genitourinary, gynecologic, head & neck, hepatobiliary, lung, melanoma, and pancreatic, were represented. Six of 28 cases (21.4%) experienced severe irAEs compared to 9/56 (16.1%) controls; the odds of developing a severe irAE were not significantly different (OR 0.43, 95% CI 0.083-2.33, p = 0.627, ns). Moreover, there were no significant differences in overall survival or tumor response between the two groups. The majority of irAEs resolved without long-term sequelae (66.7% of cases, 55.6% of controls). The majority of patients who were rechallenged with ICIs were successful in continuing therapy (66.7% of cases, 100% of controls). Conclusion Our study suggests that patients with pre-existing autoimmune disease can be treated with ICI cancer therapies and experience rates of severe irAEs and overall survival that are similar to those of the general population. These data can aid oncologists in discussing risks and benefits of ICIs when treating patients with pre-existing autoimmunity and solid tumors.
Collapse
Affiliation(s)
- Dayna Jill Isaacs
- Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Nikhita Kathuria-Prakash
- Division of Hematology and Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Robin Hilder
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Melissa G. Lechner
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Alexandra Drakaki
- Division of Hematology and Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
323
|
Klein-Brill A, Amar-Farkash S, Rosenberg-Katz K, Brenner R, Becker JC, Aran D. Comparative efficacy of combined CTLA-4 and PD-1 blockade vs. PD-1 monotherapy in metastatic melanoma: a real-world study. BJC REPORTS 2024; 2:14. [PMID: 39516682 PMCID: PMC11524072 DOI: 10.1038/s44276-024-00041-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND In light of the substantial toxicity associated with combined CTLA-4 and PD-1 blockade (ipilimumab and nivolumab), we assessed its efficacy and safety against anti-PD-1 monotherapy (nivolumab or pembrolizumab) in patients with metastatic melanoma under real-world conditions. METHODS We conducted a retrospective observational study involving 962 patients with stage IV metastatic melanoma who initiated adjuvant treatment between January 2017 and December 2021 across outpatient clinics in the United States. We adjusted for variables such as age, sex, ECOG performance status, comorbidity index, social deprivation index, metastatic sites, BRAF mutation status, and year of treatment. Outcomes included overall survival (OS) and post-treatment hospitalizations, analyzed using propensity score adjustment and inverse probability of treatment weighted Kaplan-Meier estimators. RESULTS After adjusting for all variables, no significant difference in OS was observed between treatment protocols in the overall cohort (P = 0.417). In patients with multi-organ metastasis (involvement of more than two organ systems), combined CTLA-4 and PD-1 blockade was associated with improved OS (P = 0.033). Conversely, monotherapy yielded significantly better OS in patients with oligo-organ metastasis (involvement of two or fewer organ systems; P = 0.008). Patients with oligo-organ metastasis also experienced higher hospitalization rates due to immune-related adverse events when treated with combination therapy (31.2% vs. 8.5%, P < 0.001). CONCLUSIONS Our real-world data indicate that combined CTLA-4 and PD-1 blockade is most beneficial for patients with multi-organ metastasis, while those with oligo-organ metastasis fare better with PD-1 monotherapy. The underlying reasons for these observations-whether they are due to differences in the characteristics of multi- and oligo-metastatic melanomas or the risk-benefit profile of the therapies-remain to be elucidated. These findings underscore the need for a nuanced approach to treatment regimens for stage IV melanoma patients.
Collapse
Affiliation(s)
| | | | | | - Ronen Brenner
- Department of Oncology, Edith Wolfson Medical Center, Holon, Israel
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, Essen, Germany
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), Partner Site Essen, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), University of Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dvir Aran
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
- The Taub Faculty of Computer Science, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
324
|
Akinjiyan FA, Nassief G, Phillipps J, Adeyelu T, Elliott A, Abdulla F, Zhou AY, Souroullas G, Kim KB, Vanderwalde A, Park SJ, Ansstas G. ARID2 mutations may relay a distinct subset of cutaneous melanoma patients with different outcomes. Sci Rep 2024; 14:3444. [PMID: 38341515 PMCID: PMC10858967 DOI: 10.1038/s41598-024-54136-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/08/2024] [Indexed: 02/12/2024] Open
Abstract
ARID genes encode subunits of SWI/SNF chromatin remodeling complexes and are frequently mutated in human cancers. We investigated the correlation between ARID mutations, molecular features, and clinical outcomes in melanoma patients. Cutaneous melanoma samples (n = 1577) were analyzed by next-generation sequencing. Samples were stratified by pathogenic/likely pathogenic mutation in ARID genes (ARID1A/2/1B/5B). PD-L1 expression was assessed using IHC (SP142; positive (+): ≥ 1%). Tumor mutation burden (TMB)-high was defined as ≥ 10 mutations/Mb. Transcriptomic signatures predictive of response to immune checkpoint inhibitors-interferon gamma and T-cell inflamed score were calculated. Real-world overall survival (OS) information was obtained from insurance claims data, with Kaplan-Meier estimates calculated from time of tissue collection until last date of contact. Mann-Whitney U, Chi-square, and Fisher exact tests were applied where appropriate, with p values adjusted for multiple comparisons. ARID2 mutations were more prevalent in cutaneous melanoma compared to ARID1A (11.0%: n = 451 vs 2.8%: n = 113), with concurrent ARID1A/ARID2 mutation in 1.1% (n = 46) of samples. ARID mutations were associated with a high prevalence of RAS pathway mutations-NF1 (ARID1A, 52.6%; ARID2, 48.5%; ARID1A/2, 63.6%; and ARID-WT, 13.3%; p < 0.0001) and KRAS (ARID1A, 3.5%; ARID2, 3.1%; ARID1A/2, 6.5%; and ARID-WT, 1.0%; p = 0.018)), although BRAF mutations were less common in ARID-mutated cohorts (ARID1A, 31.9%; ARID2, 35.6%; ARID1A/2, 26.1%; and ARID-WT, 50.4%; p < 0.0001). TMB-high was more common in ARID-mutated samples (ARID1A, 80.9%; ARID2, 89.9%; ARID1A/2, 100%; and ARID-WT, 49.4%; p < 0.0001), while PD-L1 positivity was similar across subgroups (ARID1A, 43.8%; ARID2, 51.1%; ARID1A/2, 52.5%; and ARID-WT, 44.9%; p = 0.109). Patients with ARID1A mutations had a higher prevalence of dMMR/MSI-H compared to those with ARID-WT (2.7% vs 0.2%, p = 0.030). Median IFN-γ and T-cell signatures were higher in ARID2-mutated samples compared to ARID-WT (IFN-γ: - 0.15 vs - 0.21, p = 0.0066; T-cell: 23.5 vs - 18.5, p = 0.041). ARID2-mutated patients had improved survival compared to ARID-WT; (HR: 1.22 (95% CI 1.0-1.5), p = 0.022). No additional OS benefit was observed with anti-PD-1 therapy for ARID2 mutation compared to ARID-WT. Melanoma patients with ARID mutations exhibited higher prevalence of markers associated with ICI response, including TMB-H, and immune-related signatures. Our data also suggests improved survival outcome in patients with ARID2 mutations, irrespective of anti-PD1 therapy.
Collapse
Affiliation(s)
- Favour A Akinjiyan
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA
| | - George Nassief
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA
| | - Jordan Phillipps
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA
| | | | | | | | - Alice Y Zhou
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA
| | - George Souroullas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA
| | - Kevin B Kim
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Soo J Park
- Division of Hematology/Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - George Ansstas
- Division of Medical Oncology, Department of Medicine, Washington University in Saint Louis, 4921 Parkview Place, Saint Louis, MO, 63110, USA.
| |
Collapse
|
325
|
Jilani S, Saco JD, Mugarza E, Pujol-Morcillo A, Chokry J, Ng C, Abril-Rodriguez G, Berger-Manerio D, Pant A, Hu J, Gupta R, Vega-Crespo A, Baselga-Carretero I, Chen JM, Shin DS, Scumpia P, Radu RA, Chen Y, Ribas A, Puig-Saus C. CAR-T cell therapy targeting surface expression of TYRP1 to treat cutaneous and rare melanoma subtypes. Nat Commun 2024; 15:1244. [PMID: 38336975 PMCID: PMC10858182 DOI: 10.1038/s41467-024-45221-2] [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: 08/17/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
A major limitation to developing chimeric antigen receptor (CAR)-T cell therapies for solid tumors is identifying surface proteins highly expressed in tumors but not in normal tissues. Here, we identify Tyrosinase Related Protein 1 (TYRP1) as a CAR-T cell therapy target to treat patients with cutaneous and rare melanoma subtypes unresponsive to immune checkpoint blockade. TYRP1 is primarily located intracellularly in the melanosomes, with a small fraction being trafficked to the cell surface via vesicular transport. We develop a highly sensitive CAR-T cell therapy that detects surface TYRP1 in tumor cells with high TYRP1 overexpression and presents antitumor activity in vitro and in vivo in murine and patient-derived cutaneous, acral and uveal melanoma models. Furthermore, no systemic or off-tumor severe toxicities are observed in an immunocompetent murine model. The efficacy and safety profile of the TYRP1 CAR-T cell therapy supports the ongoing preparation of a phase I clinical trial.
Collapse
Affiliation(s)
- Sameeha Jilani
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Justin D Saco
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Edurne Mugarza
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Aleida Pujol-Morcillo
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jeffrey Chokry
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Clement Ng
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Gabriel Abril-Rodriguez
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
| | - David Berger-Manerio
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ami Pant
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jane Hu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rubi Gupta
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Agustin Vega-Crespo
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ignacio Baselga-Carretero
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jia M Chen
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
| | - Daniel Sanghoon Shin
- Division of Hematology/Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
| | - Philip Scumpia
- Division of Dermatology, Department of Medicine, UCLA, Los Angeles, CA, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA, USA
| | - Roxana A Radu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yvonne Chen
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics at UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA
| | - Antoni Ribas
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA
| | - Cristina Puig-Saus
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA.
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA.
| |
Collapse
|
326
|
Das A, Fernandez NR, Levine A, Bianchi V, Stengs LK, Chung J, Negm L, Dimayacyac JR, Chang Y, Nobre L, Ercan AB, Sanchez-Ramirez S, Sudhaman S, Edwards M, Larouche V, Samuel D, Van Damme A, Gass D, Ziegler DS, Bielack SS, Koschmann C, Zelcer S, Yalon-Oren M, Campino GA, Sarosiek T, Nichols KE, Loret De Mola R, Bielamowicz K, Sabel M, Frojd CA, Wood MD, Glover JM, Lee YY, Vanan M, Adamski JK, Perreault S, Chamdine O, Hjort MA, Zapotocky M, Carceller F, Wright E, Fedorakova I, Lossos A, Tanaka R, Osborn M, Blumenthal DT, Aronson M, Bartels U, Huang A, Ramaswamy V, Malkin D, Shlien A, Villani A, Dirks PB, Pugh TJ, Getz G, Maruvka YE, Tsang DS, Ertl-Wagner B, Hawkins C, Bouffet E, Morgenstern DA, Tabori U. Combined Immunotherapy Improves Outcome for Replication-Repair-Deficient (RRD) High-Grade Glioma Failing Anti-PD-1 Monotherapy: A Report from the International RRD Consortium. Cancer Discov 2024; 14:258-273. [PMID: 37823831 PMCID: PMC10850948 DOI: 10.1158/2159-8290.cd-23-0559] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023]
Abstract
Immune checkpoint inhibition (ICI) is effective for replication-repair-deficient, high-grade gliomas (RRD-HGG). The clinical/biological impact of immune-directed approaches after failing ICI monotherapy is unknown. We performed an international study on 75 patients treated with anti-PD-1; 20 are progression free (median follow-up, 3.7 years). After second progression/recurrence (n = 55), continuing ICI-based salvage prolonged survival to 11.6 months (n = 38; P < 0.001), particularly for those with extreme mutation burden (P = 0.03). Delayed, sustained responses were observed, associated with changes in mutational spectra and the immune microenvironment. Response to reirradiation was explained by an absence of deleterious postradiation indel signatures (ID8). CTLA4 expression increased over time, and subsequent CTLA4 inhibition resulted in response/stable disease in 75%. RAS-MAPK-pathway inhibition led to the reinvigoration of peripheral immune and radiologic responses. Local (flare) and systemic immune adverse events were frequent (biallelic mismatch-repair deficiency > Lynch syndrome). We provide a mechanistic rationale for the sustained benefit in RRD-HGG from immune-directed/synergistic salvage therapies. Future approaches need to be tailored to patient and tumor biology. SIGNIFICANCE Hypermutant RRD-HGG are susceptible to checkpoint inhibitors beyond initial progression, leading to improved survival when reirradiation and synergistic immune/targeted agents are added. This is driven by their unique biological and immune properties, which evolve over time. Future research should focus on combinatorial regimens that increase patient survival while limiting immune toxicity. This article is featured in Selected Articles from This Issue, p. 201.
Collapse
Affiliation(s)
- Anirban Das
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatric Haematology and Oncology, Tata Medical Center, Kolkata, India
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Nicholas R. Fernandez
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Adrian Levine
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Vanessa Bianchi
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Lucie K. Stengs
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Jiil Chung
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Logine Negm
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Jose Rafael Dimayacyac
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Yuan Chang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Liana Nobre
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Ayse B. Ercan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Santiago Sanchez-Ramirez
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Sumedha Sudhaman
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Melissa Edwards
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Valerie Larouche
- Pediatric Haematology/Oncology Department, CHU de Québec-Université Laval, Quebec City, Canada
| | - David Samuel
- Department of Paediatric Oncology, Valley Children's Hospital, Madera, California
| | - An Van Damme
- Department of Paediatric Haematology and Oncology, Saint Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - David Gass
- Atrium Health/Levine Children's Hospital, Charlotte, North Carolina
| | - David S. Ziegler
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
- School of Clinical Medicine, UNSW Sydney, Sydney, Australia
| | - Stefan S. Bielack
- Department of Pediatric Oncology, Hematology and Immunology, Center for Childhood, Adolescent, and Women's Medicine, Stuttgart Cancer Center, Klinikum Stuttgart, Stuttgart, Germany
| | - Carl Koschmann
- Pediatric Hematology/Oncology, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan
| | - Shayna Zelcer
- Department of Pediatrics, London Health Sciences Centre, London, Canada
| | - Michal Yalon-Oren
- Department of Paediatric Haematology-Oncology, Sheba Medical Centre, Ramat Gan, Israel
| | - Gadi Abede Campino
- Department of Paediatric Haematology-Oncology, Sheba Medical Centre, Ramat Gan, Israel
| | | | - Kim E. Nichols
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Kevin Bielamowicz
- Department of Pediatrics, Section of Pediatric Hematology/Oncology, The University of Arkansas for Medical Sciences/Arkansas Children's Hospital, Little Rock, Arkansas
| | - Magnus Sabel
- Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg & Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Charlotta A. Frojd
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Matthew D. Wood
- Neuropathology, Oregon Health & Science University Department of Pathology, Portland, Oregon
| | - Jason M. Glover
- Department of Pediatric Hematology/Oncology, Randall Children's Hospital, Portland, Oregon
| | - Yi-Yen Lee
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Magimairajan Vanan
- Pediatric Hematology-Oncology, CancerCare Manitoba, Winnipeg, Canada
- CancerCare Manitoba Research Institute, Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Jenny K. Adamski
- Neuro-oncology Division, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Sebastien Perreault
- Neurosciences Department, Child Neurology Division, CHU Sainte-Justine, Montreal, Canada
| | - Omar Chamdine
- Pediatric Hematology Oncology, King Fahad Specialist Hospital Dammam, Eastern Province, Saudi Arabia
| | - Magnus Aasved Hjort
- Department of Paediatric Haematology and Oncology, St. Olav's University Hospital, Trondheim, Norway
| | - Michal Zapotocky
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Fernando Carceller
- Paediatric and Adolescent Neuro-Oncology and Drug Development, The Royal Marsden NHS Foundation Trust & Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Erin Wright
- Division of Neuro-Oncology, Akron Children's Hospital, Akron, Ohio
| | - Ivana Fedorakova
- Clinic of Pediatric Oncology and Hematology, University Children's Hospital, Banská Bystrica, Slovakia
| | - Alexander Lossos
- Department of Oncology, Leslie and Michael Gaffin Centre for Neuro-Oncology, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Ryuma Tanaka
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael Osborn
- Women's and Children's Hospital, North Adelaide, Australia
| | - Deborah T. Blumenthal
- Neuro-Oncology Service, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Melyssa Aronson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Canada
| | - Ute Bartels
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Annie Huang
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
| | - David Malkin
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adam Shlien
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Anita Villani
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Peter B. Dirks
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Trevor J. Pugh
- Ontario Institute for Cancer Research, Princess Margaret Cancer Centre, Toronto, Canada
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Derek S. Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Birgit Ertl-Wagner
- Department of Diagnostic Imaging, The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Daniel A. Morgenstern
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
327
|
Zhang H, Houadj L, Wu KY, Tran SD. Diagnosing and Managing Uveitis Associated with Immune Checkpoint Inhibitors: A Review. Diagnostics (Basel) 2024; 14:336. [PMID: 38337852 PMCID: PMC10855398 DOI: 10.3390/diagnostics14030336] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
This review aims to provide an understanding of the diagnostic and therapeutic challenges of uveitis associated with immune checkpoint inhibitors (ICI). In the wake of these molecules being increasingly employed as a treatment against different cancers, cases of uveitis post-ICI therapy have also been increasingly reported in the literature, warranting an extensive exploration of the clinical presentations, risk factors, and pathophysiological mechanisms of ICI-induced uveitis. This review further provides an understanding of the association between ICIs and uveitis, and assesses the efficacy of current diagnostic tools, underscoring the need for advanced techniques to enable early detection and accurate assessment. Further, it investigates the therapeutic strategies for ICI-related uveitis, weighing the benefits and limitations of existing treatment regimens, and discussing current challenges and emerging therapies in the context of their potential efficacy and side effects. Through an overview of the short-term and long-term outcomes, this article suggests recommendations and emphasizes the importance of multidisciplinary collaboration between ophthalmologists and oncologists. Finally, the review highlights promising avenues for future research and development in the field, potentially informing transformative approaches in the ocular assessment of patients under immunotherapy and the management of uveitis following ICI therapy.
Collapse
Affiliation(s)
- Huixin Zhang
- Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
| | - Lysa Houadj
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada;
| | - Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
328
|
Liu D, Wen C, Chen L, Ye M, Liu H, Sun X, Liang L, Zhang J, Chang S, Liu J. The emerging roles of PD-L1 subcellular localization in tumor immune evasion. Biochem Pharmacol 2024; 220:115984. [PMID: 38135128 DOI: 10.1016/j.bcp.2023.115984] [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: 10/07/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Targeting immune checkpoint PD-1 or its ligand PD-L1 blockade has achieved a great therapeutic effect in a variety of cancer types. However, the overall response rate and duration are still limited for intrinsic and acquired resistance. There is an urgent need to understand the underlying mechanism. Studies showed that PD-L1 regulation is related to the response to PD-1 monoclonal antibodies (PD-1 mAB). Interestingly, emerging studies found that the different distribution of PD-L1 has distinct functions in tumor through the specific signaling pathways. Thus, controlling the distribution of PD-L1 provides an attractive therapeutic strategy for enhancing PD-1 mAB efficiency and rewiring the resistance. Here, we review the recent studies about the role and regulation of PD-L1 distribution from synthesis to surface delivery, internalization, recycling, or lysosome degradation and translocated into the nucleus or secreted into the extracellular space. We place this knowledge in the context of observations in the clinic and discuss the potential therapeutic strategies to enhance the efficacy of anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan 410011, China
| | - Chengcai Wen
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan 410011, China
| | - Lu Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Hong Liu
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xing Sun
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan 410011, China
| | - Long Liang
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan 410011, China.
| | - Ji Zhang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
329
|
Di Giacomo AM, Lahn M, Eggermont AM, Fox B, Ibrahim R, Sharma P, Allison JP, Maio M. The future of targeting cytotoxic T-lymphocyte-associated protein-4: Is there a role? Eur J Cancer 2024; 198:113501. [PMID: 38169219 DOI: 10.1016/j.ejca.2023.113501] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
The 2022 yearly Think Tank Meeting in Siena, Tuscany (Italy), organized by the Italian Network for Tumor Biotherapy (NIBIT) Foundation, the Parker Institute for Cancer Immunotherapy and the World Immunotherapy Council, included a focus on the future of integrating and expanding the use of targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The conference members exchanged their views on the lessons from targeting CTLA-4 and compared the effect to the impact of blocking Programmed cell death protein 1 (PD1) or its ligand (PDL1). The increasing experience with both therapeutic approaches and their combination suggests that targeting CTLA-4 may lead to more durable responses for a sizeable proportion of patients, though the specific mechanism is not entirely understood. Overcoming toxicity of blocking CTLA-4 is currently being addressed with different doses and dose regimens, especially when combined with PD1/PDL1 blocking antibodies. Novel therapeutics targeting CTLA-4 hold the promise to reduce toxicities and thus allow different combination strategies in the future. On the whole, the consent was that targeting CTLA-4 remains an important strategy to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Anna Maria Di Giacomo
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy
| | - Michael Lahn
- IOnctura SA, Avenue Secheron 15, Geneva, Switzerland
| | - Alexander Mm Eggermont
- Princess Máxima Center and the University Medical Center Utrecht, Heidelberglaan 25, 3584 Utrecht, the Netherlands; Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximiliaan University, Munich, Germany
| | - Bernard Fox
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, 4805 NE Glisan St. Suite 2N35 Portland, OR 97213, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, 1 Letterman Drive, D3500, San Francisco, CA, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson, 1515 Holcombe Blvd, Houston, Texas 77030, USA
| | - James P Allison
- James P Allison Institute, MD Anderson, 1515 Holcombe Blvd, Texas 77030, USA
| | - Michele Maio
- University of Siena, Siena, Italy; Center for Immuno-Oncology. University Hospital of Siena, Viale Bracci, 16, Siena, Italy; NIBIT Foundation Onlus, Italy.
| |
Collapse
|
330
|
Bjursten S, Zhao Z, Al Remawi H, Studahl M, Pandita A, Simrén J, Zetterberg H, Lundell AC, Rudin A, Ny L, Levin M. Concentrations of S100B and neurofilament light chain in blood as biomarkers for checkpoint inhibitor-induced CNS inflammation. EBioMedicine 2024; 100:104955. [PMID: 38171113 PMCID: PMC10796943 DOI: 10.1016/j.ebiom.2023.104955] [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: 01/30/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Cancer treatment with immune checkpoint inhibition (ICI) can cause immune-related adverse events in the central nervous system (CNS irAE). There are no blood biomarkers to detect CNS irAE. We investigated if concentrations of S100-calcium-binding protein B (S100B) and neurofilament light chain (NfL) in blood can be used as biomarkers for CNS irAE and assessed the incidence of CNS irAE in a cohort of ICI-treated patients. METHODS In this single-centre, retrospective cohort study, we examined medical records and laboratory data of 197 consecutive patients treated with combined CTLA-4 and PD-1 inhibition (ipilimumab; ipi + nivolumab; nivo) for metastatic melanoma or renal cell carcinoma. CNS irAE was diagnosed using established criteria. Concentrations of S100B and NfL in blood were measured in patients with CNS irAE and in 84 patients without CNS irAE. FINDINGS Nine of 197 patients (4.6%) fulfilled criteria for CNS irAE. S100B and NfL in blood increased during CNS inflammation and normalized during immunosuppression. CNS irAE was detected with a sensitivity of 100% (S100B) and 79% (NfL) and a specificity of 89% (S100B) and 74% (NfL). Patients with CNS irAE had simultaneous increased concentration of C-reactive protein (CRP) (9/9) and alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) in blood (8/9). INTERPRETATION Analysis of S100B, NfL and CRP in blood facilitates the diagnosis of CNS irAE. CNS irAE may be more common than previously reported. There may be shared immune mechanisms between CNS and hepatitis irAE. FUNDING Supported by funding from the Swedish Cancer Foundation, the ALF-agreement, and Jubileumsklinikens Cancerfond.
Collapse
Affiliation(s)
- Sara Bjursten
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Zhiyuan Zhao
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hifaa Al Remawi
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marie Studahl
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ankur Pandita
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Anna-Carin Lundell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Max Levin
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
331
|
Kim TK, Han X, Hu Q, Vandsemb EN, Fielder CM, Hong J, Kim KW, Mason EF, Plowman RS, Wang J, Wang Q, Zhang JP, Badri T, Sanmamed MF, Zheng L, Zhang T, Alawa J, Lee SW, Zeidan AM, Halene S, Pillai MM, Chandhok NS, Lu J, Xu ML, Gore SD, Chen L. PD-1H/VISTA mediates immune evasion in acute myeloid leukemia. J Clin Invest 2024; 134:e164325. [PMID: 38060328 PMCID: PMC10836799 DOI: 10.1172/jci164325] [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: 08/15/2022] [Accepted: 12/06/2023] [Indexed: 02/02/2024] Open
Abstract
Acute myeloid leukemia (AML) presents a pressing medical need in that it is largely resistant to standard chemotherapy as well as modern therapeutics, such as targeted therapy and immunotherapy, including anti-programmed cell death protein (anti-PD) therapy. We demonstrate that programmed death-1 homolog (PD-1H), an immune coinhibitory molecule, is highly expressed in blasts from the bone marrow of AML patients, while normal myeloid cell subsets and T cells express PD-1H. In studies employing syngeneic and humanized AML mouse models, overexpression of PD-1H promoted the growth of AML cells, mainly by evading T cell-mediated immune responses. Importantly, ablation of AML cell-surface PD-1H by antibody blockade or genetic knockout significantly inhibited AML progression by promoting T cell activity. In addition, the genetic deletion of PD-1H from host normal myeloid cells inhibited AML progression, and the combination of PD-1H blockade with anti-PD therapy conferred a synergistic antileukemia effect. Our findings provide the basis for PD-1H as a potential therapeutic target for treating human AML.
Collapse
Affiliation(s)
- Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine
- Vanderbilt Center for Immunobiology, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
- Section of Medical Oncology
- Section of Hematology, Department of Medicine, and
| | - Xue Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Pelotonia Institute for Immuno-Oncology, OSUCCC–James Cancer Hospital
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Qianni Hu
- Division of Hematology/Oncology, Department of Medicine
| | - Esten N. Vandsemb
- Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Emily F. Mason
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - R. Skipper Plowman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Qi Wang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jian-Ping Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ti Badri
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Miguel F. Sanmamed
- Division of Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
| | - Linghua Zheng
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Pelotonia Institute for Immuno-Oncology, OSUCCC–James Cancer Hospital
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jude Alawa
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sang Won Lee
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | - Namrata S. Chandhok
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jun Lu
- Department of Genetics and
| | - Mina L. Xu
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Steven D. Gore
- Section of Hematology, Department of Medicine, and
- National Cancer Institute, Cancer Therapy Evaluation Program, Investigational Drug Branch, Bethesda, Maryland, USA
| | - Lieping Chen
- Section of Medical Oncology
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
332
|
Quail DF, Park M, Welm AL, Ekiz HA. Breast Cancer Immunity: It is TIME for the Next Chapter. Cold Spring Harb Perspect Med 2024; 14:a041324. [PMID: 37188526 PMCID: PMC10835621 DOI: 10.1101/cshperspect.a041324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Our ability to interrogate the tumor immune microenvironment (TIME) at an ever-increasing granularity has uncovered critical determinants of disease progression. Not only do we now have a better understanding of the immune response in breast cancer, but it is becoming possible to leverage key mechanisms to effectively combat this disease. Almost every component of the immune system plays a role in enabling or inhibiting breast tumor growth. Building on early seminal work showing the involvement of T cells and macrophages in controlling breast cancer progression and metastasis, single-cell genomics and spatial proteomics approaches have recently expanded our view of the TIME. In this article, we provide a detailed description of the immune response against breast cancer and examine its heterogeneity in disease subtypes. We discuss preclinical models that enable dissecting the mechanisms responsible for tumor clearance or immune evasion and draw parallels and distinctions between human disease and murine counterparts. Last, as the cancer immunology field is moving toward the analysis of the TIME at the cellular and spatial levels, we highlight key studies that revealed previously unappreciated complexity in breast cancer using these technologies. Taken together, this article summarizes what is known in breast cancer immunology through the lens of translational research and identifies future directions to improve clinical outcomes.
Collapse
Affiliation(s)
- Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Departments of Biochemistry, Oncology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce, 35430 Urla, Izmir, Turkey
| |
Collapse
|
333
|
Alhammad FA, Alburayk KB, Albadri KS, Butt SA, Azam F. Treatment response and recurrence of conjunctival melanoma with orbital invasion treated with immune checkpoint inhibitors: case report and literature review. Orbit 2024; 43:49-57. [PMID: 37052129 DOI: 10.1080/01676830.2023.2191273] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 03/05/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Conjunctival melanoma (CM) has genetic characteristics that are similar to primary cutaneous melanoma (PCM). The management of advanced CM with orbital metastasis was limited until the adoption of novel immunotherapy agents that significantly improved the survival of metastatic PCM. PURPOSE To review and compare the immune checkpoint inhibitor (ICI) treatment response in cases reported in the English literature with orbital involvement secondary to CM versus PCM. In addition, we report a case of local recurrence of CM in a young female after successful treatment with ICI. METHODS In addition to reviewing the chart of one patient who presented to our clinic, we conducted a comprehensive literature review to identify CM cases and cases with orbital metastasis secondary to advanced CM and PCM. Outcomes included patient demographics, response to ICI, and associated adverse effects. RESULTS There were ten cases with orbital involvement, four were secondary to CM, and six were metastasis from PCM. Orbital metastasis from PCM regressed following treatment with ICI agents, whereas those secondary to CM resolved completely. There were 19 cases of CM without orbital invasion. Of the 29 cases identified, complete resolution of ocular melanoma was achieved in 15 patients, representing 52% of the cases collectively, and none of them reported recurrence except in our case. CONCLUSION CM with orbital invasion responds well to ICIs, with manageable toxic effects. Despite the complete resolution, close observation is needed as the recurrence risk remains.
Collapse
Affiliation(s)
- Fatimah A Alhammad
- Oculoplastic division, Dhahran Eye Specialist Hospital, Dhahran, Saudi Arabia
| | | | - Khadija S Albadri
- Oculoplastic division, Dhahran Eye Specialist Hospital, Dhahran, Saudi Arabia
| | - Sohail A Butt
- Dammam Regional Laboratory & Blood Bank, Ministry of Health, Riyadh, Saudi Arabia
| | - Faisal Azam
- Medical Oncology Department, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| |
Collapse
|
334
|
Castro Eiro MD, Hioki K, Li L, Wilmsen MEP, Kiernan CH, Brouwers-Haspels I, van Meurs M, Zhao M, de Wit H, Grashof DGB, van de Werken HJG, Mueller YM, Schliehe C, Temizoz B, Kobiyama K, Ishii KJ, Katsikis PD. TLR9 plus STING Agonist Adjuvant Combination Induces Potent Neopeptide T Cell Immunity and Improves Immune Checkpoint Blockade Efficacy in a Tumor Model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:455-465. [PMID: 38063488 PMCID: PMC10784725 DOI: 10.4049/jimmunol.2300038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 11/10/2023] [Indexed: 01/14/2024]
Abstract
Immune checkpoint blockade (ICB) immunotherapies have emerged as promising strategies for the treatment of cancer; however, there remains a need to improve their efficacy. Determinants of ICB efficacy are the frequency of tumor mutations, the associated neoantigens, and the T cell response against them. Therefore, it is expected that neoantigen vaccinations that boost the antitumor T cell response would improve ICB therapy efficacy. The aim of this study was to develop a highly immunogenic vaccine using pattern recognition receptor agonists in combination with synthetic long peptides to induce potent neoantigen-specific T cell responses. We determined that the combination of the TLR9 agonist K-type CpG oligodeoxynucleotides (K3 CpG) with the STING agonist c-di-AMP (K3/c-di-AMP combination) significantly increased dendritic cell activation. We found that immunizing mice with 20-mer of either an OVA peptide, low-affinity OVA peptides, or neopeptides identified from mouse melanoma or lung mesothelioma, together with K3/c-di-AMP, induced potent Ag-specific T cell responses. The combined K3/c-di-AMP adjuvant formulation induced 10 times higher T cell responses against neopeptides than the TLR3 agonist polyinosinic:polycytidylic acid, a derivative of which is the leading adjuvant in clinical trials of neoantigen peptide vaccines. Moreover, we demonstrated that our K3/c-di-AMP vaccine formulation with 20-mer OVA peptide was capable of controlling tumor growth and improving survival in B16-F10-OVA tumor-bearing C57BL/6 mice and synergized with anti-PD-1 treatment. Together, our findings demonstrate that the K3/c-di-AMP vaccine formulation induces potent T cell immunity against synthetic long peptides and is a promising candidate to improve neoantigen vaccine platform.
Collapse
Affiliation(s)
- Melisa D. Castro Eiro
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kou Hioki
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ling Li
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Merel E. P. Wilmsen
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Caoimhe H. Kiernan
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Inge Brouwers-Haspels
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marjan van Meurs
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Manzhi Zhao
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Harm de Wit
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dwin G. B. Grashof
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Yvonne M. Mueller
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Christopher Schliehe
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Burcu Temizoz
- Division of Vaccine Science, Department of Microbiology and Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ken J. Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Peter D. Katsikis
- Department of Immunology; Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
335
|
Zhou S, Zhang S, Zheng K, Li Z, Hu E, Mu Y, Mai J, Zhao A, Zhao Z, Li F. Salmonella-mediated methionine deprivation drives immune activation and enhances immune checkpoint blockade therapy in melanoma. J Immunother Cancer 2024; 12:e008238. [PMID: 38302417 PMCID: PMC10836381 DOI: 10.1136/jitc-2023-008238] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Although immune checkpoint inhibitor (ICI)-based therapy is advantageous for patients with advanced melanoma, resistance and relapse are frequent. Thus, it is crucial to identify effective drug combinations and develop new therapies for the treatment of melanoma. SGN1, a genetically modified Salmonella typhimurium species that causes the targeted deprivation of methionine in tumor tissues, is currently under investigation in clinical trials. However, the inhibitory effect of SGN1 on melanoma and the benefits of SGN1 in combination with ICIs remain largely unexplored. Therefore, this study aims to investigate the antitumor potential of SGN1, and its ability to enhance the efficacy of antibody-based programmed cell death-ligand 1 (PD-L1) inhibitors in the treatment of murine melanoma. METHODS The antitumor activity of SGN1 and the effect of SGN1 on the efficacy of PD-L1 inhibitors was studied through murine melanoma models. Further, The Cancer Genome Atlas-melanoma cohort was clustered using ConsensusClusterPlus based on the methionine deprivation-related genes, and immune characterization was performed using xCell, Microenvironment Cell Populations-counter, Estimation of Stromal and Immune cells in MAlignant Tumor tissues using Expression data, and immunophenoscore (IPS) analyses. The messenger RNA data on programmed death-1 (PD-1) immunotherapy response were obtained from the Gene Expression Omnibus database. Gene Set Enrichment Analysis of methionine deprivation-up gene set was performed to determine the differences between pretreatment responders and non-responders. RESULTS This study showed that both, the intratumoral and the intravenous administration of SGN1 in subcutaneous B16-F10 melanomas, suppress tumor growth, which was associated with an activated CD8+T-cell response in the tumor microenvironment. Combination therapy of SGN1 with systemic anti-PD-L1 therapy resulted in better antitumor activity than the individual monotherapies, respectively, and the high therapeutic efficacy of the combination was associated with an increase in the systemic level of tumor-specific CD8+ T cells. Two clusters consisting of methionine deprivation-related genes were identified. Patients in cluster 2 had higher expression of methionine_deprivation_up genes, better clinical outcomes, and higher immune infiltration levels compared with patients in cluster 1. Western blot, IPS analysis, and immunotherapy cohort study revealed that methionine deficiency may show a better response to ICI therapy CONCLUSIONS:: This study reports Salmonella-based SGN1 as a potent anticancer agent against melanoma, and lays the groundwork for the potential synergistic effect of ICIs and SGN1 brought about by improving the immune microenvironment in melanomas.
Collapse
Affiliation(s)
- Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Shiwei Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Kexin Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zixuan Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Enyu Hu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Jialuo Mai
- Guangzhou Sinogen Pharmaceutical Co Ltd, Guangzhou, Guangdong, China
| | - Allan Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
336
|
Dailah HG, Hommdi AA, Koriri MD, Algathlan EM, Mohan S. Potential role of immunotherapy and targeted therapy in the treatment of cancer: A contemporary nursing practice. Heliyon 2024; 10:e24559. [PMID: 38298714 PMCID: PMC10828696 DOI: 10.1016/j.heliyon.2024.e24559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Immunotherapy and targeted therapy have emerged as promising therapeutic options for cancer patients. Immunotherapies induce a host immune response that mediates long-lived tumor destruction, while targeted therapies suppress molecular mechanisms that are important for tumor maintenance and growth. In addition, cytotoxic agents and targeted therapies regulate immune responses, which increases the chances that these therapeutic approaches may be efficiently combined with immunotherapy to ameliorate clinical outcomes. Various studies have suggested that combinations of therapies that target different stages of anti-tumor immunity may be synergistic, which can lead to potent and more prolonged responses that can achieve long-lasting tumor destruction. Nurses associated with cancer patients should have a better understanding of the immunotherapies and targeted therapies, such as their efficacy profiles, mechanisms of action, as well as management and prophylaxis of adverse events. Indeed, this knowledge will be important in establishing care for cancer patients receiving immunotherapies and targeted therapies for cancer treatment. Moreover, nurses need a better understanding regarding targeted therapies and immunotherapies to ameliorate outcomes in patients receiving these therapies, as well as management and early detection of possible adverse effects, especially adverse events associated with checkpoint inhibitors and various other therapies that control T-cell activation causing autoimmune toxicity. Nurses practice in numerous settings, such as hospitals, home healthcare agencies, radiation therapy facilities, ambulatory care clinics, and community agencies. Therefore, as compared to other members of the healthcare team, nurses often have better opportunities to develop the essential rapport in providing effective nurse-led patient education, which is important for effective therapeutic outcomes and continuance of therapy. In this article, we have particularly focused on providing a detailed overview on targeted therapies and immunotherapies used in cancer treatment, management of their associated adverse events, and the impact as well as strategies of nurse-led patient education.
Collapse
Affiliation(s)
- Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Abdullah Abdu Hommdi
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Mahdi Dafer Koriri
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Essa Mohammed Algathlan
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| |
Collapse
|
337
|
Wang H, Yang J, Li X, Zhao H. Current state of immune checkpoints therapy for glioblastoma. Heliyon 2024; 10:e24729. [PMID: 38298707 PMCID: PMC10828821 DOI: 10.1016/j.heliyon.2024.e24729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Glioblastoma (GBM), one of the most aggressive forms of brain cancer, has limited treatment options. Recent years have witnessed the remarkable success of checkpoint inhibitor immunotherapy across various cancer types. Against this backdrop, several clinical trials investigating checkpoint inhibitors for GBM are underway in multiple countries. Furthermore, the integration of immunotherapy with traditional treatment approaches is now emerging as a highly promising strategy. This review summarizes the latest advancements in checkpoint inhibitor immunotherapy for GBM treatment. We provide a concise yet comprehensive overview of current GBM immunotherapy options. Additionally, this review underscores combination strategies and potential biomarkers for predicting response and resistance in GBM immunotherapies.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Jing Yang
- Department of Emergency Surgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| | - Xiangjun Li
- School of medicine, Department of Breast surgery, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, Shandong, 266005, China
| |
Collapse
|
338
|
Smith A, Boby JM, Benny SJ, Ghazali N, Vermeulen E, George M. Immunotherapy in Older Patients with Cancer: A Narrative Review. Int J Gen Med 2024; 17:305-313. [PMID: 38298248 PMCID: PMC10830099 DOI: 10.2147/ijgm.s435001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Purpose Immunotherapies have revolutionized cancer treatment; however, relatively little is known about their efficacy and toxicity in the elderly, a cohort accounting for more than half of total cancer cases. In this review, we aim to provide insight into the current knowledge base regarding the clinical utility and side effects of immunotherapies in the geriatric population as well as identify key gaps in the literature where further research is essential. Methods We conducted a rapid critical review of available literature, focusing on studies reporting on use of immunotherapy in cancer patients aged ≥65 years. The review assessed studies that included different types of cancer, were of multiple study types (although predominantly retrospective), had different study duration, and reported different outcomes of interest. Owing to this heterogeneity, meta-analysis and a direct comparison between studies were not feasible. Results Overall, the review findings indicate that certain malignancies have shown comparable survival rates in younger and older age groups when managed with immunotherapeutic drugs, the incidence of immunotherapy-related side effects varies only slightly by age groups, and in general there is a lack of studies on the determinants of the clinical outcomes of immunotherapy in or including geriatric patients. Conclusion Enhanced clinical benefits along with better tolerability associated with immunotherapies make it an attractive alternative to conventional chemotherapeutic drugs, especially in elderly patients. There is currently a limited number of studies assessing the clinical outcomes of immunotherapies, particularly in the elderly. Overall, our findings reflect a need for further prospective studies focussing on geriatric patients representative of the real-life population, in order to derive a more precise understanding of the clinical utility, toxicity profile, and cost-effectiveness of immune checkpoint inhibitors in older patients with cancer.
Collapse
Affiliation(s)
- Alexandra Smith
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| | | | | | | | - Elke Vermeulen
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| | - Mathew George
- Tamworth Hospital, Hunter New England Local Health District (NSW Health), Tamworth, NSW, Australia
| |
Collapse
|
339
|
Luan J, Zhang D, Liu B, Yang A, Lv K, Hu P, Yu H, Shmuel A, Zhang C, Ma G. Immune-related lncRNAs signature and radiomics signature predict the prognosis and immune microenvironment of glioblastoma multiforme. J Transl Med 2024; 22:107. [PMID: 38279111 PMCID: PMC10821572 DOI: 10.1186/s12967-023-04823-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 12/22/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults. This study aimed to construct immune-related long non-coding RNAs (lncRNAs) signature and radiomics signature to probe the prognosis and immune infiltration of GBM patients. METHODS We downloaded GBM RNA-seq data and clinical information from The Cancer Genome Atlas (TCGA) project database, and MRI data were obtained from The Cancer Imaging Archive (TCIA). Then, we conducted a cox regression analysis to establish the immune-related lncRNAs signature and radiomics signature. Afterward, we employed a gene set enrichment analysis (GSEA) to explore the biological processes and pathways. Besides, we used CIBERSORT to estimate the abundance of tumor-infiltrating immune cells (TIICs). Furthermore, we investigated the relationship between the immune-related lncRNAs signature, radiomics signature and immune checkpoint genes. Finally, we constructed a multifactors prognostic model and compared it with the clinical prognostic model. RESULTS We identified four immune-related lncRNAs and two radiomics features, which show the ability to stratify patients into high-risk and low-risk groups with significantly different survival rates. The risk score curves and Kaplan-Meier curves confirmed that the immune-related lncRNAs signature and radiomics signature were a novel independent prognostic factor in GBM patients. The GSEA suggested that the immune-related lncRNAs signature were involved in L1 cell adhesion molecular (L1CAM) interactions and the radiomics signature were involved signaling by Robo receptors. Besides, the two signatures was associated with the infiltration of immune cells. Furthermore, they were linked with the expression of critical immune genes and could predict immunotherapy's clinical response. Finally, the area under the curve (AUC) (0.890,0.887) and C-index (0.737,0.817) of the multifactors prognostic model were greater than those of the clinical prognostic model in both the training and validation sets, indicated significantly improved discrimination. CONCLUSIONS We identified the immune-related lncRNAs signature and tradiomics signature that can predict the outcomes, immune cell infiltration, and immunotherapy response in patients with GBM.
Collapse
Affiliation(s)
- Jixin Luan
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Di Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kuan Lv
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Pianpian Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Hongwei Yu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Chuanchen Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China.
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China.
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
340
|
Li C, Chen S, Fang X, Du Y, Guan XY, Lin R, Xu L, Lan P, Yan Q. LOXL1 promotes tumor cell malignancy and restricts CD8 + T cell infiltration in colorectal cancer. Cell Biol Toxicol 2024; 40:6. [PMID: 38267662 PMCID: PMC10808464 DOI: 10.1007/s10565-024-09840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer mortality globally. Lymph node metastasis and immunosuppression are main factors of poor prognosis in CRC patients. Lysyl oxidase like 1 (LOXL1), part of the lysyl oxidase (LOX) family, plays a yet unclear role in CRC. This study aimed to identify effective biomarkers predictive of prognosis and efficacy of immunotherapy in CRC patients, and to elucidate the prognostic value, clinical relevance, functional and molecular features, and immunotherapy predictive role of LOXL1 in CRC and pan-cancer. METHODS Weighted gene co-expression network analysis (WGCNA) was employed to explore gene modules related to tumor metastasis and CD8 + T cell infiltration. LOXL1 emerged as a hub gene through differential gene expression and survival analysis. The molecular signatures, functional roles, and immunological characteristics affected by LOXL1 were analyzed in multiple CRC cohorts, cell lines and clinical specimens. Additionally, LOXL1's potential as an immunotherapy response indicator was assessed, along with its role in pan-cancer. RESULTS Turquoise module in WGCNA analysis was identified as the hub module associated with lymph node metastasis and CD8 + T cell infiltration. Aberrant elevated LOXL1 expression was observed in CRC and correlated with poorer differentiation status and prognosis. Molecular and immunological characterization found that LOXL1 might mediate epithelial-mesenchymal transition (EMT) process and immunosuppressive phenotypes of CRC. Functional study found that LOXL1 enhanced tumor cell proliferation, migration and invasion. Moreover, high LOXL1 levels corresponded to reduced CD8 + T cell infiltration and predicted poor clinical outcomes of immunotherapy. Similar trends were also observed at the pan-cancer level. CONCLUSIONS Our findings underscore the critical role of LOXL1 in modulating both malignancy and immunosuppression in CRC. This positions LOXL1 as a promising biomarker for predicting prognosis and the response to immunotherapy in CRC patients.
Collapse
Affiliation(s)
- Chenxi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
| | - Siqi Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China
| | - Xiaona Fang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yaqing Du
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Liang Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China.
| | - Qian Yan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Room 703, Building No. 3, 26 Yuancun ERheng Road, Guangzhou, 510655, China.
| |
Collapse
|
341
|
Zhang J, Ren Z, Hu Y, Shang S, Wang R, Ma J, Zhang Z, Wu M, Wang F, Yu J, Chen D. High HPK1 +PD-1 +TIM-3 +CD8 + T cells infiltration predicts poor prognosis to immunotherapy in NSCLC patients. Int Immunopharmacol 2024; 127:111363. [PMID: 38101218 DOI: 10.1016/j.intimp.2023.111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/21/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
At present the efficacy of immune checkpoint inhibitors (ICIs) remains limited. The lack of responsiveness in certain patients may be attributed to CD8+ T cell exhaustion within the tumor microenvironment (TME). Hematopoietic progenitor kinase 1 (HPK1) has been identified as a mediator of T cell dysfunction, leading to our hypothesis that HPK1 positive exhausted CD8+ T cells could serve as a predictor for ICIs' efficacy in NSCLC patients, and potentially indicate key cellular subset causing ICIs resistance. Here, we retrospectively collected tumor tissue samples from 36 NSCLC patients who underwent first-line immunotherapy. Using multiplex immunohistochemistry, we visualized various PD-1+CD8+ T cell subsets and explore biomarkers for response. The analysis endpoints included overall response rate (ORR), progression free survival (PFS), and overall survival (OS), correlating them with levels of cell infiltration or effective density. We found that the proportion of PD-1+CD8+ T cell subsets did not align with predictions for ORR, PFS, and OS. Conversely, a high infiltration of HPK1+PD-1+TIM-3+CD8+ T cells was identified as an independent risk factor for both PFS (P = 0.019) and OS (P = 0.03). These cells were found to express the highest levels of Granzyme B, and the secretion of Granzyme B in CD8+ T cell subsets was related to TCF-1. In conclusion, these data suggest that a high infiltration of HPK1+PD-1+TIM-3+CD8+ T cells correlates with poor clinical outcomes in NSCLC patients receiving immunotherapy. These cells may represent terminally exhausted T cells that fail to respond to ICIs, thereby laying the groundwork for the potential integration of HPK1 inhibitors with immunotherapy to enhance treatment strategy.
Collapse
Affiliation(s)
- Jingxin Zhang
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziyuan Ren
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shijie Shang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruiyang Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiachun Ma
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zengfu Zhang
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fei Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Dawei Chen
- Shandong University Cancer Center, Jinan, Shandong, China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
342
|
Sorrentino FS, De Rosa F, Di Terlizzi P, Toneatto G, Gabai A, Finocchio L, Salati C, Spadea L, Zeppieri M. Uveal melanoma: Recent advances in immunotherapy. World J Clin Oncol 2024; 15:23-31. [PMID: 38292657 PMCID: PMC10823941 DOI: 10.5306/wjco.v15.i1.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular cancer in adults. The incidence in Europe and the United States is 6-7 per million population per year. Although most primary UMs can be successfully treated and locally controlled by irradiation therapy or local tumor resection, up to 50% of UM patients develop metastases that usually involve the liver and are fatal within 1 year. To date, chemotherapy and targeted treatments only obtain minimal responses in patients with metastatic UM, which is still characterized by poor prognosis. No standard therapeutic approaches for its prevention or treatment have been established. The application of immunotherapy agents, such as immune checkpoint inhibitors that are effective in cutaneous melanoma, has shown limited effects in the treatment of ocular disease. This is due to UM's distinct genetics, natural history, and complex interaction with the immune system. Unlike cutaneous melanomas characterized mainly by BRAF or NRAS mutations, UMs are usually triggered by a mutation in GNAQ or GNA11. As a result, more effective immunotherapeutic approaches, such as cancer vaccines, adoptive cell transfer, and other new molecules are currently being studied. In this review, we examine novel immunotherapeutic strategies in clinical and preclinical studies and highlight the latest insight in immunotherapy and the development of tailored treatment of UM.
Collapse
Affiliation(s)
| | - Francesco De Rosa
- Department of Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, Meldola 47014, Italy
| | - Patrick Di Terlizzi
- Department of Surgical Sciences, Unit of Ophthalmology, Ospedale Maggiore, Bologna 40100, Italy
| | - Giacomo Toneatto
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Andrea Gabai
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Lucia Finocchio
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, “Sapienza” University of Rome, Rome 00142, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| |
Collapse
|
343
|
Santry LA, van Vloten JP, AuYeung AWK, Mould RC, Yates JGE, McAusland TM, Petrik JJ, Major PP, Bridle BW, Wootton SK. Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer. Front Microbiol 2024; 15:1325558. [PMID: 38328418 PMCID: PMC10847535 DOI: 10.3389/fmicb.2024.1325558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024] Open
Abstract
Introduction Tumor microenvironments are immunosuppressive due to progressive accumulation of mutations in cancer cells that can drive expression of a range of inhibitory ligands and cytokines, and recruitment of immunomodulatory cells, including myeloid-derived suppressor cells (MDSC), tumor-associated macrophages, and regulatory T cells (Tregs). Methods To reverse this immunosuppression, we engineered mesogenic Newcastle disease virus (NDV) to express immunological checkpoint inhibitors anti-cytotoxic T lymphocyte antigen-4 and soluble programmed death protein-1. Results Intratumoral administration of recombinant NDV (rNDV) to mice bearing intradermal B16-F10 melanomas or subcutaneous CT26LacZ colon carcinomas led to significant changes in the tumor-infiltrating lymphocyte profiles. Vectorizing immunological checkpoint inhibitors in NDV increased activation of intratumoral natural killer cells and cytotoxic T cells and decreased Tregs and MDSCs, suggesting induction of a pro-inflammatory state with greater infiltration of activated CD8+ T cells. These notable changes translated to higher ratios of activated effector/suppressor tumor-infiltrating lymphocytes in both cancer models, which is a promising prognostic marker. Whereas all rNDV-treated groups showed evidence of tumor regression and increased survival in the CT26LacZ and B16-F10, only treatment with NDV expressing immunological checkpoint blockades led to complete responses compared to tumors treated with NDV only. Discussion These data demonstrated that NDV expressing immunological checkpoint inhibitors could reverse the immunosuppressive state of tumor microenvironments and enhance tumor-specific T cell responses.
Collapse
Affiliation(s)
- Lisa A. Santry
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob P. van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Amanda W. K. AuYeung
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Robert C. Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jacob G. E. Yates
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Thomas M. McAusland
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - James J. Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
344
|
Gu XY, Huo JL, Yu ZY, Jiang JC, Xu YX, Zhao LJ. Immunotherapy in hepatocellular carcinoma: an overview of immune checkpoint inhibitors, drug resistance, and adverse effects. ONCOLOGIE 2024; 26:9-25. [DOI: 10.1515/oncologie-2023-0412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Hepatocellular carcinoma (HCC) is a concerning liver cancer with rising incidence and mortality rates worldwide. The effectiveness of traditional therapies in managing advanced HCC is limited, necessitating the development of new therapeutic strategies. Immune checkpoint inhibitors (ICIs) have emerged as a promising strategy for HCC management. By preventing tumor cells from evading immune surveillance through immunological checkpoints, ICIs can restore the immune system’s ability to target and eliminate tumors. While ICIs show promise in enhancing the immune response against malignancies, challenges such as drug resistance and adverse reactions hinder their efficacy. To address these challenges, developing individualized ICI treatment strategies is critical. Combining targeted therapy and immunotherapy holds the potential for comprehensive therapeutic effects. Additionally, biomarker-based individualized ICI treatment strategies offer promise in predicting treatment response and guiding personalized patient care. Future research should explore emerging ICI treatment methods to optimize HCC immunotherapy. This review provides an overview of ICIs as a new treatment for HCC, demonstrating some success in promoting the tumor immune response. However, drug resistance and adverse reactions remain important considerations that must be addressed. As tailored treatment plans evolve, the prospect of immunotherapy for HCC is expected to grow, offering new opportunities for improved patient outcomes.
Collapse
Affiliation(s)
- Xuan-Yu Gu
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| | - Jin-Long Huo
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| | - Zhi-Yong Yu
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| | - Ji-Chang Jiang
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| | - Ya-Xuan Xu
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| | - Li-Jin Zhao
- Department of General Surgery , Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University , Zunyi , China
| |
Collapse
|
345
|
Shahab SW, Patil P, Fangusaro JR, Patteson B, Goldman-Yassen A, Eaton BR, Boydston W, Schniederjan M, Aguilera D. Primary Diffuse Leptomeningeal Melanomatosis in a Child with Extracranial Metastasis: Case Report. Curr Oncol 2024; 31:579-587. [PMID: 38275834 PMCID: PMC10814890 DOI: 10.3390/curroncol31010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Primary meningeal melanomatosis is an extremely rare tumor with very few documented responses to treatment. A 3-year-old male with a complex past medical history, including prematurity and shunted hydrocephalus, was diagnosed with primary meningeal melanomatosis with peritoneal implants. Molecular testing revealed an NRAS Q61R mutation. The patient received proton craniospinal radiation followed by immunotherapy with nivolumab (1 mg/kg) and ipilimumab (3 mg/kg) IV every 3 weeks and, upon progression, he was switched to a higher dose of nivolumab (3 mg/kg IV every 2 weeks) and binimetinib (24 mg/m2/dose, twice a day). The patient had significant improvement of CNS disease with radiation therapy and initial immunotherapy but progression of extracranial metastatic peritoneal and abdominal disease. Radiation was not administered to the whole abdomen. After two cycles of nivolumab and treatment with the MEK inhibitor binimetinib, he had radiographic and clinical improvement in abdominal metastasis and ascitis. He ultimately died from RSV infection, Klebsiella sepsis, and subdural hemorrhage without evidence of tumor progression. This is the first report of a child with primary meningeal melanomatosis with extracranial metastatic disease with response to a combination of radiation, immunotherapy and MEK inhibitor therapy.
Collapse
Affiliation(s)
- Shubin W. Shahab
- Aflac Cancer and Blood Disorders Center, Atlanta, GA 30342, USA; (J.R.F.); (B.P.); (D.A.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Prabhumallikarjun Patil
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jason R. Fangusaro
- Aflac Cancer and Blood Disorders Center, Atlanta, GA 30342, USA; (J.R.F.); (B.P.); (D.A.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Brooke Patteson
- Aflac Cancer and Blood Disorders Center, Atlanta, GA 30342, USA; (J.R.F.); (B.P.); (D.A.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
| | - Adam Goldman-Yassen
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Radiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bree R. Eaton
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Winship Cancer Institute, Atlanta, GA 30322, USA
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William Boydston
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Matthew Schniederjan
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dolly Aguilera
- Aflac Cancer and Blood Disorders Center, Atlanta, GA 30342, USA; (J.R.F.); (B.P.); (D.A.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA; (P.P.); (A.G.-Y.); (B.R.E.); (W.B.); (M.S.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
346
|
Adhikary S, Pathak S, Palani V, Acar A, Banerjee A, Al-Dewik NI, Essa MM, Mohammed SGAA, Qoronfleh MW. Current Technologies and Future Perspectives in Immunotherapy towards a Clinical Oncology Approach. Biomedicines 2024; 12:217. [PMID: 38255322 PMCID: PMC10813720 DOI: 10.3390/biomedicines12010217] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Immunotherapy is now established as a potent therapeutic paradigm engendering antitumor immune response against a wide range of malignancies and other diseases by modulating the immune system either through the stimulation or suppression of immune components such as CD4+ T cells, CD8+ T cells, B cells, monocytes, macrophages, dendritic cells, and natural killer cells. By targeting several immune checkpoint inhibitors or blockers (e.g., PD-1, PD-L1, PD-L2, CTLA-4, LAG3, and TIM-3) expressed on the surface of immune cells, several monoclonal antibodies and polyclonal antibodies have been developed and already translated clinically. In addition, natural killer cell-based, dendritic cell-based, and CAR T cell therapies have been also shown to be promising and effective immunotherapeutic approaches. In particular, CAR T cell therapy has benefited from advancements in CRISPR-Cas9 genome editing technology, allowing the generation of several modified CAR T cells with enhanced antitumor immunity. However, the emerging SARS-CoV-2 infection could hijack a patient's immune system by releasing pro-inflammatory interleukins and cytokines such as IL-1β, IL-2, IL-6, and IL-10, and IFN-γ and TNF-α, respectively, which can further promote neutrophil extravasation and the vasodilation of blood vessels. Despite the significant development of advanced immunotherapeutic technologies, after a certain period of treatment, cancer relapses due to the development of resistance to immunotherapy. Resistance may be primary (where tumor cells do not respond to the treatment), or secondary or acquired immune resistance (where tumor cells develop resistance gradually to ICIs therapy). In this context, this review aims to address the existing immunotherapeutic technologies against cancer and the resistance mechanisms against immunotherapeutic drugs, and explain the impact of COVID-19 on cancer treatment. In addition, we will discuss what will be the future implementation of these strategies against cancer drug resistance. Finally, we will emphasize the practical steps to lay the groundwork for enlightened policy for intervention and resource allocation to care for cancer patients.
Collapse
Affiliation(s)
- Subhamay Adhikary
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Vignesh Palani
- Faculty of Medicine, Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, 06800 Ankara, Türkiye;
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Nader I. Al-Dewik
- Department of Pediatrics, Women’s Wellness and Research Center, Hamad Medical Corporation, Doha 00974, Qatar;
| | - Musthafa Mohamed Essa
- College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat 123, Oman
| | | | - M. Walid Qoronfleh
- Research & Policy Division, Q3 Research Institute (QRI), Ypsilanti, MI 48917, USA
| |
Collapse
|
347
|
Filderman JN, Taylor JL, Wang J, Zhang Y, Singh P, Ross MA, Watkins SC, Nedal Al Bzour A, Karapetyan L, Kalinski P, Storkus WJ. Antagonism of regulatory ISGs enhances the anti-melanoma efficacy of STING agonists. Front Immunol 2024; 15:1334769. [PMID: 38312842 PMCID: PMC10835797 DOI: 10.3389/fimmu.2024.1334769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
Background Stimulator of Interferon Genes (STING) is a dsDNA sensor that triggers type I inflammatory responses. Recent data from our group and others support the therapeutic efficacy of STING agonists applied intratumorally or systemically in a range of murine tumor models, with treatment benefits associated with tumor vascular normalization and improved immune cell recruitment and function within the tumor microenvironment (TME). However, such interventions are rarely curative and STING agonism coordinately upregulates expression of immunoregulatory interferon-stimulated genes (ISGs) including Arg2, Cox2, Isg15, Nos2, and Pdl1 that may limit treatment benefits. We hypothesized that combined treatment of melanoma-bearing mice with STING agonist ADU-S100 together with antagonists of regulatory ISGs would result in improved control of tumor growth vs. treatment with ADU-S100 alone. Methods Mice bearing either B16 (BRAFWTPTENWT) or BPR20 (BRAFV600EPTEN-/-) melanomas were treated with STING agonist ADU-S100 plus various inhibitors of ARG2, COX2, NOS2, PD-L1, or ISG15. Tumor growth control and changes in the TME were evaluated for combination treatment vs ADU-S100 monotherapy by tumor area measurements and flow cytometry/transcriptional profiling, respectively. Results In the B16 melanoma model, we noted improved antitumor efficacy only when ADU-S100 was combined with neutralizing/blocking antibodies against PD-L1 or ISG15, but not inhibitors of ARG2, COX2, or NOS2. Conversely, in the BPR20 melanoma model, improved tumor growth control vs. ADU-S100 monotherapy was only observed when combining ADU-S100 with ARG2i, COX2i, and NOS2i, but not anti-PD-L1 or anti-ISG15. Immune changes in the TME associated with improved treatment outcomes were subtle but included increases in proinflammatory innate immune cells and activated CD8+CD69+ T cells and varied between the two tumor models. Conclusions These data suggest contextual differences in the relative contributions of individual regulatory ISGs that serve to operationally limit the anti-tumor efficacy of STING agonists which should be considered in future design of novel combination protocols for optimal treatment benefit.
Collapse
Affiliation(s)
- Jessica N Filderman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Yali Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Prashant Singh
- Genomics Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark A Ross
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ayah Nedal Al Bzour
- Department of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
348
|
Shen H, Mullen L, Ojo OA, Xing C, Yassin A, Lewis Z, Bonner JA, Shi LZ. HIF1α-glycolysis engages activation-induced cell death to drive IFN-γ induction in hypoxic T cells. RESEARCH SQUARE 2024:rs.3.rs-3830704. [PMID: 38260594 PMCID: PMC10802708 DOI: 10.21203/rs.3.rs-3830704/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The role of HIF1α-glycolysis in regulating IFN-γ induction in hypoxic T cells is unknown. Given that hypoxia is a common feature in a wide array of pathophysiological contexts such as tumor and that IFN-γ is instrumental for protective immunity, it is of great significance to gain a clear idea on this. Combining pharmacological and genetic gain-of-function and loss-of-function approaches, we find that HIF1α-glycolysis controls IFN-γ induction in both human and mouse T cells activated under hypoxia. Specific deletion of HIF1α in T cells (HIF1α-/-) and glycolytic inhibition significantly abrogate IFN-γ induction. Conversely, HIF1α stabilization in T cells by hypoxia and VHL deletion (VHL-/-) promotes IFN-γ production. Mechanistically, reduced IFN-γ production in hypoxic HIF1α-/- T cells is due to attenuated activation-induced cell death but not proliferative defect. We further show that depletion of intracellular acetyl-CoA is a key metabolic underlying mechanism. Hypoxic HIF1α-/- T cells are less able to kill tumor cells, and HIF1α-/- tumor-bearing mice are not responsive to immune checkpoint blockade (ICB) therapy, indicating loss of HIF1α in T cells is a major mechanism of therapeutic resistance to ICBs. Importantly, acetate supplementation restores IFN-γ production in hypoxic HIF1α-/- T cells and re-sensitizes HIF1α-/- tumor-bearing mice to ICBs, providing an effective strategy to overcome ICB resistance. Taken together, our results highlight T cell HIF1α-anaerobic glycolysis as a principal mediator of IFN-γ induction and anti-tumor immunity. Considering that acetate supplementation (i.e., glycerol triacetate (GTA)) is approved to treat infants with Canavan disease, we envision a rapid translation of our findings, justifying further testing of GTA as a repurposed medicine for ICB resistance, a pressing unmet medical need.
Collapse
Affiliation(s)
- Hongxing Shen
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Logan Mullen
- Genomics Core Laboratory, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, 99775, USA
| | - Oluwagbemiga A. Ojo
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Chuan Xing
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Abdelrahman Yassin
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - Zach Lewis
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
| | - James A. Bonner
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, UAB-SOM, USA
| | - Lewis Zhichang Shi
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, UAB-SOM, USA
- Department of Microbiology and Immunology Institute, UAB-SOM, USA
- Department of Pharmacology and Toxicology, UAB-SOM, USA
- Immunology Institute, UAB-SOM, USA
| |
Collapse
|
349
|
Liu L, van Schaik TA, Chen KS, Rossignoli F, Borges P, Vrbanac V, Wakimoto H, Shah K. Establishment and immune phenotyping of patient-derived glioblastoma models in humanized mice. Front Immunol 2024; 14:1324618. [PMID: 38274817 PMCID: PMC10808686 DOI: 10.3389/fimmu.2023.1324618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive and common type of malignant brain tumor diagnosed in adults. Preclinical immunocompetent mouse tumor models generated using mouse tumor cells play a pivotal role in testing the therapeutic efficacy of emerging immune-based therapies for GBMs. However, the clinical translatability of such studies is limited as mouse tumor lines do not fully recapitulate GBMs seen in inpatient settings. In this study, we generated three distinct, imageable human-GBM (hGBM) models in humanized mice using patient-derived GBM cells that cover phenotypic and genetic GBM heterogeneity in primary (invasive and nodular) and recurrent tumors. We developed a pipeline to first enrich the tumor-initiating stem-like cells and then successfully established robust patient-derived GBM tumor engraftment and growth in bone marrow-liver-thymus (BLT) humanized mice. Multiplex immunofluorescence of GBM tumor sections revealed distinct phenotypic features of the patient GBM tumors, with myeloid cells dominating the immune landscape. Utilizing flow cytometry and correlative immunofluorescence, we profiled the immune microenvironment within the established human GBM tumors in the BLT mouse models and showed tumor infiltration of variable human immune cells, creating a unique immune landscape compared with lymphoid organs. These findings contribute substantially to our understanding of GBM biology within the context of the human immune system in humanized mice and lay the groundwork for further translational studies aimed at advancing therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Longsha Liu
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Thijs A. van Schaik
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Kok-Siong Chen
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Filippo Rossignoli
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Paulo Borges
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Vladimir Vrbanac
- Humanized Immune System Mouse Program, Ragon Institute, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Hiroaki Wakimoto
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
| |
Collapse
|
350
|
Li Y, Pond G, McWhirter E. Multisystem Immune-Related Adverse Events from Dual-Agent Immunotherapy Use. Curr Oncol 2024; 31:425-435. [PMID: 38248113 PMCID: PMC10813982 DOI: 10.3390/curroncol31010028] [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: 10/17/2023] [Revised: 11/27/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND little is known about the incidence and characteristics of multisystem immune-related adverse events (irAEs) associated with dual-agent ipilimumab and nivolumab use. METHODS A retrospective cohort review was completed that included cancer patients seen at the Juravinski Cancer Centre who received at least one dose of ipilimumab and nivolumab from 2018 to 2022. Patient characteristics, cancer types, and irAEs were recorded. Multivariate logistic and cox regressions were completed, comparing those who developed multisystem irAEs, single irAE, and no irAE. RESULTS A total of 123 patients were included in this study. Out of 123 patients, 72 (59%) had melanoma, 50/123 (41%) had renal cell carcinoma (RCC), and 1/123 (1%) had breast cancer. Multisystem irAEs were seen in 40% of the overall cohort. The most common irAE type was dermatitis (22%), followed by colitis (19%) and hepatitis (17%). CONCLUSIONS Our study demonstrated that multisystem irAEs are prevalent amongst patients receiving ipilimumab and nivolumab. It is important for both physician education and the counseling and consent of patients to monitor the potential for multiple irAEs.
Collapse
Affiliation(s)
- Yuchen Li
- Department of Oncology, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Gregory Pond
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Elaine McWhirter
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| |
Collapse
|