1
|
Shen C, Zhang J, Zhang G, Luo H, Zhang S, Yuan Q, Xu W, Wang W, Miao L. Research hotspots and trends in the field of immune checkpoint inhibitors (ICIs) for cervical cancer: A bibliometric study from 2014 to 2024. Hum Vaccin Immunother 2025; 21:2483031. [PMID: 40159879 PMCID: PMC11959921 DOI: 10.1080/21645515.2025.2483031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/11/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
In recent years, immune checkpoint inhibitors (ICIs) have emerged as a novel immunotherapeutic approach, offering renewed hope for enhancing cervical cancer patient prognosis. This study represents the inaugural bibliometric analysis of ICIs in the context of cervical cancer, covering the period from 2014 to 2024. A total of 422 articles were identified through the Web of Science Core Collection database, amassing 10,977 citations, with a consistent annual increase in the number of publications. The leading contributors in terms of countries, institutions, journals, and authors included China, the University of Texas System, Frontiers in Oncology, and Bradley J. Monk, respectively. The journal with the highest frequency of citation and co-citation was Journal of Clinical Oncology. The researchers with the highest number of citations and co-citations were Sarina A Piha-Paul and Krishnansu S Tewari respectively. The keyword cluster analysis identified four main research directions. Furthermore, literature co-citation analysis and burst citation analysis revealed three research hotspots and four potential emerging topics within this domain, respectively. This study provides valuable reference and enlightenment for researchers in this field. As research progresses, ICIs are anticipated to offer significant hope and breakthroughs in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Chaoyan Shen
- Department of Ultrasound, Jining NO.1 People’s Hospital, Jining, China
| | - Juan Zhang
- Department of Hematology, Jining NO.1 people’ Hospital, Jining, China
| | - Guanyu Zhang
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Hao Luo
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Shuangke Zhang
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Quan Yuan
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Wei Xu
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Wei Wang
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| | - Lele Miao
- Department of Thyroid and Breast Surgery, Jining NO.1 people’ Hospital, Jining, China
| |
Collapse
|
2
|
Reschke R, Enk AH, Hassel JC. Prognostic Biomarkers in Evolving Melanoma Immunotherapy. Am J Clin Dermatol 2025; 26:213-223. [PMID: 39707058 PMCID: PMC11850490 DOI: 10.1007/s40257-024-00910-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Melanoma, a highly aggressive form of skin cancer, has seen significant advancements in treatment through the introduction of immunotherapy. However, the variability in patient responses underscores the need for reliable biomarkers to guide treatment decisions. This article reviews key biomarkers in melanoma immunotherapy, such as PD-L1 expression, tumor mutational burden (TMB), and gene expression profiles (GEPs). It also explores emerging biomarkers, including LAG-3 expression, immune cell phenotyping in tissue and blood, gut microbiota, and circulating tumor DNA (ctDNA). Notably, ctDNA may offer valuable insights into the efficacy of T cell-engaging bispecific molecules, such as tebentafusp. The review provides a comprehensive overview of the evolving landscape of melanoma biomarkers, their role in personalizing treatment, and future research directions, including neoadjuvant immune checkpoint inhibition.
Collapse
Affiliation(s)
- Robin Reschke
- Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), Heidelberg University, NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, 69120, Heidelberg, Germany.
| | - Alexander H Enk
- Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), Heidelberg University, NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Medical Faculty Heidelberg, Department of Dermatology and National Center for Tumor Diseases (NCT), Heidelberg University, NCT Heidelberg, a partnership between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, 69120, Heidelberg, Germany
| |
Collapse
|
3
|
Zielińska MK, Ciążyńska M, Sulejczak D, Rutkowski P, Czarnecka AM. Mechanisms of Resistance to Anti-PD-1 Immunotherapy in Melanoma and Strategies to Overcome It. Biomolecules 2025; 15:269. [PMID: 40001572 PMCID: PMC11853485 DOI: 10.3390/biom15020269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/22/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025] Open
Abstract
Resistance to anti-PD-1 therapy in melanoma remains a major obstacle in achieving effective and durable treatment outcomes, highlighting the need to understand and address the underlying mechanisms. The first key factor is innate anti-PD-1 resistance signature (IPRES), an expression of a group of genes associated with tumor plasticity and immune evasion. IPRES promotes epithelial-to-mesenchymal transition (EMT), increasing melanoma cells' invasiveness and survival. Overexpressed AXL, TWIST2, and WNT5a induce phenotypic changes. The upregulation of pro-inflammatory cytokines frequently coincides with EMT-related changes, further promoting a resistant and aggressive tumor phenotype. Inflamed tumor microenvironment may also drive the expression of resistance. The complexity of immune resistance development suggests that combination therapies are necessary to overcome it. Furthermore, targeting epigenetic regulation and exploring novel approaches such as miR-146a modulation may provide new strategies to counter resistance in melanoma.
Collapse
Affiliation(s)
- Magdalena K. Zielińska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Faculty of Medicine, Warsaw Medical University, 02-091 Warsaw, Poland
| | - Magdalena Ciążyńska
- Chemotherapy Unit and Day Chemotherapy Ward, Specialised Oncology Hospital, 97-200 Tomaszów Mazowiecki, Poland;
- Department of Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, 91-347 Łódź, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.K.Z.); (P.R.)
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
4
|
Li WJ, Najdawi W, Badla O, Galor A, Karp CL. Immune Checkpoint Inhibitors in the Treatment of Ocular Surface Cancers: A Review. Semin Ophthalmol 2025:1-11. [PMID: 39923258 DOI: 10.1080/08820538.2025.2458658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/01/2025] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed cancer therapy by targeting key immune pathways such as PD-1, PD-L1, CTLA-4, and LAG-3 to enhance the immune system's ability to combat malignancies. Their use in treating ocular surface tumors is an emerging area of interest, particularly in conjunctival melanoma (CM) and ocular surface squamous neoplasia (OSSN). Some studies have indicated the potential of ICI's in sebaceous gland carcinoma (SeC), conjunctival lymphoma, and Kaposi sarcoma. PURPOSE This review aims to evaluate the role of ICIs in treating ocular surface tumors, focusing on their mechanisms of action, clinical outcomes, and therapeutic potential. METHODS A literature review was conducted by searching Pubmed for studies published between January 2014 and October 2024. Studies included were original research, clinical trials, case reports and series, and reviews. RESULTS ICIs, including pembrolizumab and nivolumab, have shown promising results in CM, achieving tumor regression and disease stabilization in advanced and metastatic cases. ICIs have also demonstrated efficacy in OSSN, particularly in lesions with high tumor mutational burden, with responses ranging from partial to complete resolution. Although clinical data for SeC and conjunctival lymphoma remain limited to isolated reports, these studies suggest a role for ICIs in managing refractory or advanced disease. CONCLUSION ICIs hold transformative potential in improving outcomes for ocular surface malignancies, particularly in cases where conventional treatments fail or pose significant morbidity. Despite their promise, challenges persist, including variable response rates, immune-related adverse events, and the need for reliable predictive biomarkers. Comprehensive prospective studies are necessary to refine the application of ICIs, optimize treatment strategies, and expand therapeutic options for these challenging cancers.
Collapse
Affiliation(s)
- Wendy J Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Wisam Najdawi
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Omar Badla
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
- Department of Ophthalmology, Miami Veterans Hospital, Miami, FL, USA
| | - Carol L Karp
- Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, FL, USA
| |
Collapse
|
5
|
Mareboina M, Bakhl K, Agioti S, Yee NS, Georgakopoulos-Soares I, Zaravinos A. Comprehensive Analysis of Granzymes and Perforin Family Genes in Multiple Cancers. Biomedicines 2025; 13:408. [PMID: 40002821 PMCID: PMC11853441 DOI: 10.3390/biomedicines13020408] [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: 11/05/2024] [Revised: 01/25/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Cancer remains a significant global health concern, with immunotherapies emerging as promising treatments. This study explored the role of perforin-1 (PRF1) and granzymes A, B and K (GZMA, GZMB and GZMK) in cancer biology, focusing on their impact on tumor cell death and immune response modulation. Methods: Through a comprehensive genomic analysis across various cancer types, we explored the differential expression, mutation profiles and methylation patterns of these genes, providing insights into their potential as therapeutic targets. Furthermore, we investigated their association with immune cell infiltration and pathway activation within the tumor microenvironment in each tumor type. Results: Our findings revealed distinct expression patterns and prognostic implications for PRF1, GZMA, GZMB and GZMK across different cancers, highlighting their multifaceted roles in tumor immunity. We found increased immune infiltration across all tumor types and significant correlations between the genes of interest and cytotoxic T cells, as well as the most significant survival outcomes in breast cancer. We also show that granzymes and perforin-1 are significantly associated with indicators of immunosuppression and T cell dysfunction within patient cohorts. In skin melanoma, glioblastoma, kidney and bladder cancers, we found significant correlations between the genes of interest and patient survival after receiving immune-checkpoint inhibition therapy. Additionally, we identified potential associations between the mRNA expression levels of these genes and drug sensitivity. Conclusions: Overall, this study enhances our understanding of the molecular mechanisms underlying tumor immunity and provides valuable insights into the potential therapeutic implications of PRF1, GZMA, GZMB and GZMK in cancer treatment.
Collapse
Affiliation(s)
- Manvita Mareboina
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Katrina Bakhl
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Stephanie Agioti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Nelson S. Yee
- Department of Medicine, Division of Hematology-Oncology, Penn State Health Milton S. Hershey Medical Center, Next-Generation Therapies Program, Penn State Cancer Institute, Hershey, PA 17033, USA;
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.M.); (K.B.)
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| |
Collapse
|
6
|
Lezcano C, Luo L, Shen R, Orlow I, Thomas NE, Berwick M, Busam KJ. Solar elastosis correlates with high tumor mutation burden and better 5-year disease-specific survival in patients with stage II/III melanoma. EJC SKIN CANCER 2024; 2:100274. [PMID: 39801746 PMCID: PMC11721741 DOI: 10.1016/j.ejcskn.2024.100274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Objective To evaluate the relation between solar elastosis and tumor mutation burden (TMB) in a large clinically annotated cohort of stage II and III melanoma patients. Methods Primary cutaneous melanomas from 469 AJCC (8th edition) stage II and III patients with clinical annotation including outcome at 5 years of diagnosis were histopathologically evaluated for solar elastosis. Next-generation sequencing assay MSK-IMPACT™ was employed to determine TMB. Analysis by Fisher's exact test, chi-square, and Kruskal-Wallis were performed, as well as uni- and multivariate logistic regression. Results Tumors stratified by low and high TMB showed marked and statistically significant differences in presence and extent of associated solar elastosis. Lower risk patient stage (II versus III by AJCC 8th edition) as well as better 5-year melanomaspecific survival (as binary variable of controls-survivors versus cases-dead of disease at 5 years of diagnosis) were associated with severe solar elastosis. On univariate and multivariate logistic regression models, severe solar elastosis predicted significantly decreased odds of dying of melanoma within 5 years of diagnosis (OR 0.60, 95 % CI 0.39-0.89; and OR 0.42, 95 % CI 0.20-0.83, respectively; both p<0.05). Conclusion The association of solar elastosis to TMB and 5-year melanoma specific survival points to its potential as a biomarker of clinical relevance that can be assessed by routine histopathology.
Collapse
Affiliation(s)
- Cecilia Lezcano
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Li Luo
- Department of Internal Medicine and the University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Ronglai Shen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy E. Thomas
- Department of Dermatology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Marianne Berwick
- Department of Internal Medicine and the University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Klaus J. Busam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
7
|
Chhabra Y, Fane ME, Pramod S, Hüser L, Zabransky DJ, Wang V, Dixit A, Zhao R, Kumah E, Brezka ML, Truskowski K, Nandi A, Marino-Bravante GE, Carey AE, Gour N, Maranto DA, Rocha MR, Harper EI, Ruiz J, Lipson EJ, Jaffee EM, Bibee K, Sunshine JC, Ji H, Weeraratna AT. Sex-dependent effects in the aged melanoma tumor microenvironment influence invasion and resistance to targeted therapy. Cell 2024; 187:6016-6034.e25. [PMID: 39243764 PMCID: PMC11580838 DOI: 10.1016/j.cell.2024.08.013] [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: 01/23/2023] [Revised: 02/19/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
There is documented sex disparity in cutaneous melanoma incidence and mortality, increasing disproportionately with age and in the male sex. However, the underlying mechanisms remain unclear. While biological sex differences and inherent immune response variability have been assessed in tumor cells, the role of the tumor-surrounding microenvironment, contextually in aging, has been overlooked. Here, we show that skin fibroblasts undergo age-mediated, sex-dependent changes in their proliferation, senescence, ROS levels, and stress response. We find that aged male fibroblasts selectively drive an invasive, therapy-resistant phenotype in melanoma cells and promote metastasis in aged male mice by increasing AXL expression. Intrinsic aging in male fibroblasts mediated by EZH2 decline increases BMP2 secretion, which in turn drives the slower-cycling, highly invasive, and therapy-resistant melanoma cell phenotype, characteristic of the aged male TME. Inhibition of BMP2 activity blocks the emergence of invasive phenotypes and sensitizes melanoma cells to BRAF/MEK inhibition.
Collapse
Affiliation(s)
- Yash Chhabra
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | - Mitchell E Fane
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Sneha Pramod
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laura Hüser
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Daniel J Zabransky
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Agrani Dixit
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ruzhang Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Edwin Kumah
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Megan L Brezka
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kevin Truskowski
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Asmita Nandi
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gloria E Marino-Bravante
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Naina Gour
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Devon A Maranto
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Murilo R Rocha
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth I Harper
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Justin Ruiz
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Evan J Lipson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA; The Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kristin Bibee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joel C Sunshine
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
8
|
Özgü E, Kaplan BG, Sivakumar S, Sokol ES, Aydın E, Tokat ÜM, Adibi A, Karakoç EG, Hu J, Kurzrock R, Demiray M. Therapeutic vulnerabilities and pan-cancer landscape of BRAF class III mutations in epithelial solid tumors. BJC REPORTS 2024; 2:77. [PMID: 39516363 PMCID: PMC11524077 DOI: 10.1038/s44276-024-00086-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Kinase-impaired class III BRAF mutations have recently received attention as a possible prognostic factor and therapeutic target. Class III BRAF variants differ from class I and class II mutations in terms of mechanism of pathway activation and therapeutic vulnerabilities. Genomic landscape analyses of tumors in large real-world cohorts represent a great opportunity to further characterize tumor-related molecular events and treatment vulnerabilities, however, such data is not yet available for tumors with BRAF class III mutations. METHODS We investigated the pan-cancer genomic landscape of BRAF class III mutations in 376,302 patients. Patients had comprehensive genomic profiling either by FoundationOne® or FoundationOne®CDx from formalin-fixed, paraffin embedded tissue biopsies. 2 patient cases that harbored BRAF class III mutations who demonstrated dramatic response to anti-EGFR treatment were presented. RESULTS BRAF class III mutations are likely to co-occur with RAF1, NRAS and HRAS alterations, while concomitant KRAS alterations were rare. Moreover, we found that alterations that predict resistance to anti-EGFR agents were significantly less common in tumors harboring BRAF class III mutations, which is of great importance as anti-EGFR therapies are a potential targeted treatment option in these tumors. DISCUSSION Our findings suggest a heterogenous interplay of oncogenic alterations in BRAF class III mutated tumors and have important implications for the molecular mechanisms of carcinogenesis while revealing potential therapeutic vulnerabilities. HIGHLIGHTS Tumors harboring BRAF class III (BRAF vIII) mutations comprise a novel subset with distinct genomic heterogeneity. BRAF vIII mutations may sensitize tumors to anti-EGFR treatments. BRAF vIII alterations show significantly less co-occurrence with alterations that predict resistance to anti-EGFR agents. Rare tumors with limited therapy options should be screened for BRAF vIII mutations as they may benefit from anti-EGFR agents.
Collapse
Affiliation(s)
- Eylül Özgü
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey
| | | | | | | | - Esranur Aydın
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey
| | - Ünal Metin Tokat
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey
| | - Ashkan Adibi
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey
| | - Ebru Gül Karakoç
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey
| | - Jiancheng Hu
- National Cancer Center Singapore, Division of Cellular and Molecular Research, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS National Cancer Centre, 8 College Road, 169857, Singapore, Singapore
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, USA
- WIN Consortium, Paris, France
| | - Mutlu Demiray
- Medicana International Atasehir Hospital, Demiray Precision Oncology Center, Istanbul, Turkey.
| |
Collapse
|
9
|
Taherifard E, Tran K, Saeed A, Yasin JA, Saeed A. Biomarkers for Immunotherapy Efficacy in Advanced Hepatocellular Carcinoma: A Comprehensive Review. Diagnostics (Basel) 2024; 14:2054. [PMID: 39335733 PMCID: PMC11431712 DOI: 10.3390/diagnostics14182054] [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: 07/26/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver malignancy and the sixth most common cancer globally, remains fatal for many patients with inappropriate responses to treatment. Recent advancements in immunotherapy have transformed the treatment landscape for advanced HCC. However, variability in patient responses to immunotherapy highlights the need for biomarkers that can predict treatment outcomes. This manuscript comprehensively reviews the evolving role of biomarkers in immunotherapy efficacy, spanning from blood-derived indicators-alpha-fetoprotein, inflammatory markers, cytokines, circulating tumor cells, and their DNA-to tissue-derived indicators-programmed cell death ligand 1 expression, tumor mutational burden, microsatellite instability, and tumor-infiltrating lymphocytes. The current body of evidence suggests that these biomarkers hold promise for improving patient selection and predicting immunotherapy outcomes. Each biomarker offers unique insights into disease biology and the immune landscape of HCC, potentially enhancing the precision of treatment strategies. However, challenges such as methodological variability, high costs, inconsistent findings, and the need for large-scale validation in well-powered two-arm trial studies persist, making them currently unsuitable for integration into standard care. Addressing these challenges through standardized techniques and implementation of further studies will be critical for the future incorporation of these biomarkers into clinical practice for advanced HCC.
Collapse
Affiliation(s)
- Erfan Taherifard
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Krystal Tran
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Ali Saeed
- Department of Medicine, Ochsner Lafayette General Medical Center, Lafayette, LA 70503, USA
| | - Jehad Amer Yasin
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
10
|
Mihaila RI, Gheorghe AS, Zob DL, Stanculeanu DL. A Complete Response in a Metastatic Melanoma Patient After a Single Dose of Dual Checkpoint Inhibitors Blockade Could Be Predictable: A Case Report. Cureus 2024; 16:e69301. [PMID: 39282490 PMCID: PMC11393513 DOI: 10.7759/cureus.69301] [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] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Cutaneous malignant melanoma is one of the most aggressive forms of skin cancer and thus, a high mortality has been reported over decades. The prognosis for melanoma varies widely based on several factors, including the stage at which it is diagnosed, the location and thickness of the tumor, the patient's age and overall health, and specific genetic factors associated with melanoma. Therapeutic options include checkpoint inhibitors, regardless of V-Raf Murine Sarcoma Viral Oncogene Homolog B status (BRAF), and targeted therapy (anti-BRAF) in the adjuvant or metastatic setting. Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment but predicting which patients will benefit from these therapies remains challenging. Biomarkers like leukocytes, neutrophils, eosinophils, basophils, platelets, and other peripheral blood biomarkers have been investigated for their potential to predict responses to ICIs. Tumor mutational burden (TMB), circulating tumor DNA (ctDNA), and soluble PD-L1 (sPD-L1) have emerged as potential biomarkers for predicting responses to ICIs. Elevated baseline levels of ctDNA and elevated sPD-L1 levels have been associated with worse prognosis in melanoma patients. High TMB is often associated with better responses to ICIs in melanoma. Here we present a case from our department, of a 57-year-old patient, diagnosed in 2019 with stage IV - pT4cNx cM1 (lymph nodes metastases) and suspicion of lung metastases, BRAF wild-type right hallux malignant melanoma. Due to impressive results, first-line treatment with ICIs nivolumab and ipilimumab was the preferred treatment of choice, which showed a favorable response, with regression of oncological disease after the first cycle, and achieving complete response afterward. Unfortunately, the treatment was discontinued due to severe hepatic and pancreatic toxicity, but the favorable response to immunotherapy has been maintained for four years and is ongoing. Identifying predictive biomarkers is important to achieve the best response for the patient, with minimal adverse events, especially if long-term clinical benefit can be reached.
Collapse
Affiliation(s)
- Raluca Ioana Mihaila
- Department of Oncology, University of Medicine and Pharmacy Bucharest, Bucharest, ROU
- Department of Medical Oncology I, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", Bucharest, ROU
| | - Adelina Silvana Gheorghe
- Department of Medical Oncology I, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", Bucharest, ROU
| | - Daniela Luminita Zob
- Department of Medical Oncology II, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", Bucharest, ROU
| | - Dana Lucia Stanculeanu
- Department of Medical Oncology, Institute of Oncology "Prof. Dr. Alexandru Trestioreanu", Bucharest, ROU
| |
Collapse
|
11
|
Reveneau MF, Masliah-Planchon J, Fernandez M, Ouikene A, Dron B, Dadamessi I, Dayen C, Golmard L, Chauffert B. Major response of a peritoneal mesothelioma to nivolumab and ipilimumab: a case report, molecular analysis and review of literature. Front Oncol 2024; 14:1410322. [PMID: 39091916 PMCID: PMC11291227 DOI: 10.3389/fonc.2024.1410322] [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: 03/31/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Malignant peritoneal mesothelioma (MPM) is a rare tumor associated with a poor prognosis and a lack of consensus regarding treatment strategies. While the Checkmate 743 trial demonstrated the superiority of first-line nivolumab and ipilimumab over chemotherapy in malignant pleural mesothelioma (MPlM), few studies have assessed the effectiveness of immunotherapy against MPM, due to its rarity. Here, we report a major and sustained 12-month response in a 74-year-old female patient who received the anti-PD-1 nivolumab and the anti-CTLA4 ipilimumab as first-line therapy for diffuse MPM. PD-L1 was expressed and BAP1 expression was lost, as shown by immunohistochemistry, however the BAP1 gene was not mutated. Our findings suggest a role for ICI in non-resectable diffuse MPM exhibiting PD-L1 overexpression and loss of BAP1 expression, and instill new hope in their treatment. To our knowledge, this is the second reported case of dual immunotherapy used as first-line in MPM with a major clinical response. To investigate the clinical outcome, we conducted additional molecular analyses of the MPM tumor and we reviewed the literature on immunotherapy in MPM to discuss the role of PD-L1 and BAP1.
Collapse
Affiliation(s)
- Marie-Florence Reveneau
- Department of Genetics, Institut Curie, Paris, France
- Department of Medical Oncology, Saint Quentin Hospital, Saint Quentin, France
| | | | - Manuel Fernandez
- Department of Radiology, Saint Quentin Hospital, Saint Quentin, France
| | - Abdenour Ouikene
- Department of Medical Oncology, Saint Quentin Hospital, Saint Quentin, France
| | - Bernard Dron
- Department of Digestive Surgery, Saint Quentin Hospital, Saint Quentin, France
| | - Innocenti Dadamessi
- Department of Digestive Surgery, Saint Quentin Hospital, Saint Quentin, France
| | - Charles Dayen
- Department of Pneumology, Saint Quentin Hospital, Saint Quentin, France
| | - Lisa Golmard
- Department of Genetics, Institut Curie, Paris, France
| | - Bruno Chauffert
- Department of Medical Oncology, Saint Quentin Hospital, Saint Quentin, France
| |
Collapse
|
12
|
Rakhmilevich AL, Tsarovsky NW, Felder M, Zaborek J, Moram S, Erbe AK, Pieper AA, Spiegelman DV, Cheng EM, Witt CM, Overwijk WW, Morris ZS, Sondel PM. A combined radio-immunotherapy regimen eradicates late-stage tumors in mice. Front Immunol 2024; 15:1419773. [PMID: 39076988 PMCID: PMC11284032 DOI: 10.3389/fimmu.2024.1419773] [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: 04/18/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Background The majority of experimental approaches for cancer immunotherapy are tested against relatively small tumors in tumor-bearing mice, because in most cases advanced cancers are resistant to the treatments. In this study, we asked if even late-stage mouse tumors can be eradicated by a rationally designed combined radio-immunotherapy (CRI) regimen. Methods CRI consisted of local radiotherapy, intratumoral IL-12, slow-release systemic IL-2 and anti- CTLA-4 antibody. Therapeutic effects of CRI against several weakly immunogenic and immunogenic mouse tumors including B78 melanoma, MC38 and CT26 colon carcinomas and 9464D neuroblastoma were evaluated. Immune cell depletion and flow cytometric analysis were performed to determine the mechanisms of the antitumor effects. Results Tumors with volumes of 2,000 mm3 or larger were eradicated by CRI. Flow analyses of the tumors revealed reduction of T regulatory (Treg) cells and increase of CD8/Treg ratios following CRI. Rapid shrinkage of the treated tumors did not require T cells, whereas T cells were involved in the systemic effect against the distant tumors. Cured mice developed immunological memory. Conclusions These findings underscore that rationally designed combination immunotherapy regimens can be effective even against large, late-stage tumors.
Collapse
Affiliation(s)
| | - Noah W. Tsarovsky
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Mildred Felder
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Jen Zaborek
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Sritha Moram
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Alexander A. Pieper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Dan V. Spiegelman
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Emily M. Cheng
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Cole M. Witt
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
13
|
Sorino C, Iezzi S, Ciuffreda L, Falcone I. Immunotherapy in melanoma: advances, pitfalls, and future perspectives. Front Mol Biosci 2024; 11:1403021. [PMID: 39086722 PMCID: PMC11289331 DOI: 10.3389/fmolb.2024.1403021] [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: 03/18/2024] [Accepted: 05/16/2024] [Indexed: 08/02/2024] Open
Abstract
Cutaneous melanoma is the deadliest and most aggressive form of skin cancer owing to its high capacity for metastasis. Over the past few decades, the management of this type of malignancy has undergone a significant revolution with the advent of both targeted therapies and immunotherapy, which have greatly improved patient quality of life and survival. Nevertheless, the response rates are still unsatisfactory for the presence of side effects and development of resistance mechanisms. In this context, tumor microenvironment has emerged as a factor affecting the responsiveness and efficacy of immunotherapy, and the study of its interplay with the immune system has offered new promising clinical strategies. This review provides a brief overview of the currently available immunotherapeutic strategies for melanoma treatment by analyzing both the positive aspects and those that require further improvement. Indeed, a better understanding of the mechanisms involved in the immune evasion of melanoma cells, with particular attention on the role of the tumor microenvironment, could provide the basis for improving current therapies and identifying new predictive biomarkers.
Collapse
|
14
|
Goodman RS, Jung S, Fletcher K, Burnette H, Mohyuddin I, Irlmeier R, Ye F, Johnson DB. Primary Tumor Characteristics as Biomarkers of Immunotherapy Response in Advanced Melanoma: A Retrospective Cohort Study. Cancers (Basel) 2024; 16:2355. [PMID: 39001417 PMCID: PMC11240575 DOI: 10.3390/cancers16132355] [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: 05/27/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Identifying patients likely to benefit from immune checkpoint inhibitor (ICI) treatment remains a crucial goal for melanoma. The objective of this study is to assess the association between primary tumor features and immunotherapy response and survival in advanced melanoma patients. In this single-center retrospective cohort study, disease characteristics, response to immunotherapy, PFS, and OS were assessed among melanoma patients (excluding mucosal and uveal primaries) treated with ICI. Among 447 patients, 300 (67.1%) received anti-PD-1 monotherapy and 147 (32.9%) received ipilimumab/nivolumab. A total of 338 (75.6%) had cutaneous melanoma, 29 (6.5%) had acral melanoma, and 80 (17.9%) had melanoma of unknown primary. Ulceration and stage at initial presentation were associated with inferior outcomes on univariate analysis. However, on multivariate analysis, this result was not observed, but cutaneous melanoma and each of its subtypes (superficial spreading, nodular, other, unknown) were positively associated with response, longer PFS, and longer OS. Metastatic stage (M1c, M1d) at presentation (OR = 1.8, p < 0.05) and BRAFV600E mutation status (OR = 1.6, p < 0.001) were associated with shorter PFS. This study is limited by its retrospective and single-center design. Cutaneous melanoma and its subtypes were significantly associated with response, PFS, and OS compared with acral or unknown primary melanoma.
Collapse
Affiliation(s)
- Rachel S Goodman
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Seungyeon Jung
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Kylie Fletcher
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Hannah Burnette
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | | | - Rebecca Irlmeier
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Fei Ye
- Vanderbilt University School of Medicine, Nashville, TN 37240, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Douglas B Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| |
Collapse
|
15
|
Szász I, Koroknai V, Várvölgyi T, Pál L, Szűcs S, Pikó P, Emri G, Janka E, Szabó IL, Ádány R, Balázs M. Identification of Plasma Lipid Alterations Associated with Melanoma Metastasis. Int J Mol Sci 2024; 25:4251. [PMID: 38673837 PMCID: PMC11050015 DOI: 10.3390/ijms25084251] [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: 03/18/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The aim of this study was to apply a state-of-the-art quantitative lipidomic profiling platform to uncover lipid alterations predictive of melanoma progression. Our study included 151 melanoma patients; of these, 83 were without metastasis and 68 with metastases. Plasma samples were analyzed using a targeted Lipidyzer™ platform, covering 13 lipid classes and over 1100 lipid species. Following quality control filters, 802 lipid species were included in the subsequent analyses. Total plasma lipid contents were significantly reduced in patients with metastasis. Specifically, levels of two out of the thirteen lipid classes (free fatty acids (FFAs) and lactosylceramides (LCERs)) were significantly decreased in patients with metastasis. Three lipids (CE(12:0), FFA(24:1), and TAG47:2-FA16:1) were identified as more effective predictors of melanoma metastasis than the well-known markers LDH and S100B. Furthermore, the predictive value substantially improved upon combining the lipid markers. We observed an increase in the cumulative levels of five lysophosphatidylcholines (LPC(16:0); LPC(18:0); LPC(18:1); LPC(18:2); LPC(20:4)), each individually associated with an elevated risk of lymph node metastasis but not cutaneous or distant metastasis. Additionally, seventeen lipid molecules were linked to patient survival, four of which (CE(12:0), CE(14:0), CE(15:0), SM(14:0)) overlapped with the lipid panel predicting metastasis. This study represents the first comprehensive investigation of the plasma lipidome of melanoma patients to date. Our findings suggest that plasma lipid profiles may serve as important biomarkers for predicting clinical outcomes of melanoma patients, including the presence of metastasis, and may also serve as indicators of patient survival.
Collapse
Affiliation(s)
- István Szász
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (R.Á.)
| | - Viktória Koroknai
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| | - Tünde Várvölgyi
- Department of Dermatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (G.E.); (E.J.); (I.L.S.)
| | - László Pál
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| | - Sándor Szűcs
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| | - Péter Pikó
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (G.E.); (E.J.); (I.L.S.)
| | - Eszter Janka
- Department of Dermatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (G.E.); (E.J.); (I.L.S.)
| | - Imre Lőrinc Szabó
- Department of Dermatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.V.); (G.E.); (E.J.); (I.L.S.)
| | - Róza Ádány
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (R.Á.)
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| | - Margit Balázs
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (I.S.); (R.Á.)
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4028 Debrecen, Hungary; (V.K.); (L.P.); (S.S.); (P.P.)
| |
Collapse
|
16
|
Gleber-Netto FO, Nagarajan P, Sagiv O, Pickering CR, Gross N, Ning J, Yeshi MM, Mitku Y, Tetzlaff MT, Esmaeli B. Histologic and Genomic Analysis of Conjunctival SCC in African and American Cohorts Reveal UV Light and HPV Signatures and High Tumor Mutation Burden. Invest Ophthalmol Vis Sci 2024; 65:24. [PMID: 38597722 PMCID: PMC11008748 DOI: 10.1167/iovs.65.4.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/12/2023] [Indexed: 04/11/2024] Open
Abstract
Purpose Conjunctival squamous cell carcinoma (conjSCC) is more prevalent and aggressive in sub-Saharan African countries compared with the rest of the world. This study aims to compare the genomic, immunophenotypic, and histologic features between patients from the United States and Ethiopia, to identify etiopathogenic mechanisms and unveil potential treatment strategies. Methods We compared histologic features and mutational profiles using whole exome sequencing, high-risk human papillomavirus (HPV) status, PD-L1 expression, and tumor-infiltrating lymphocytes in conjSCC tumors of patients from Ethiopia (ETH; n = 25) and the United States (from MD Anderson [the MDA cohort]; n = 29). Genomic alterations were compared with SCCs from other anatomic sites using data from The Cancer Genome Atlas. Results Solar elastosis was seen in 78% of ETH and 10% of MDA samples. Thicker tumors had higher density of CD8+ and CD3+ cells. HPV status was similar between the cohorts (ETH = 21% and MDA = 28%). The mean tumor mutation burden (TMB) was significantly higher in conjSCC (3.01/Mb, log10) and cutaneous SCC compared other SCC subtypes. ETH samples had higher TMB compared to the MDA cohort (3.34 vs. 2.73). Mutations in genes associated with ultraviolet light (UV) signature were most frequently encountered (SBS7b = 74% and SBS7a = 72%), with higher prevalence in the ETH cohort, whereas SBS2 and SBS13 signatures were more common among MDA HPV+ conjSCCs. Conclusions Our findings suggest that UV exposure may play a major role in conjSCC, with a higher prevalence in the ETH cohort compared with the MDA cohort, where HPV also contributes.
Collapse
Affiliation(s)
- Frederico O. Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Priyadharsini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Oded Sagiv
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
- The Goldschleger Eye Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Curtis R. Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Neil Gross
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Yonas Mitku
- Department of Ophthalmology, Mekelle University, Mekelle, Ethiopia
| | - Michael T. Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Bita Esmaeli
- Orbital Oncology and Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| |
Collapse
|
17
|
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
|
18
|
Ma C, Teng Q, Shang L, Du F, Li L. Tumor mutation load better predicts the prognosis of patients treated with immune checkpoint inhibitors in upper gastrointestinal cancers: A systematic review and meta-analysis. Cancer Rep (Hoboken) 2024; 7:e1959. [PMID: 38204354 PMCID: PMC10849990 DOI: 10.1002/cnr2.1959] [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: 08/10/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Tumor mutational load (TML) has emerged as a potential biomarker for multiple solid tumors. However, data on its prognostic impact on upper gastrointestinal (UGI) cancer are limited. Therefore, the aim of this systematic review and meta-analysis was to assess the prognostic value of TML for the survival of patients with UGI cancer. METHOD A comprehensive search of the PubMed, Embase, Cochrane Library, and Web of Science databases was conducted up to February 13, 2023. Eleven studies met our inclusion criteria. Hazard ratios (HRs) for progression-free survival and overall survival and their 95% confidence intervals (CIs) were calculated. Subsequently, the combined HR and its 95% CI were calculated for UGI tract cancers in the high and low TML groups. I2 statistics and p-values were used to evaluate heterogeneity. Publication bias, sensitivity, and subgroup analyses were performed to determine sources of heterogeneity. RESULTS In total, 932 patients with UGI tract cancer from 11 publications were included. The high TML group treated with immunotherapy showed significantly improved overall survival (HR = 0.68; 95% CI: 0.53, 0.86; p = .001) and progression-free survival (HR = 0.74; 95% CI: 0.58, 0.95; p = .020) compared with the low TML group. CONCLUSION Our study demonstrated that patients with UGI tumors and higher TML have a better prognosis with immunotherapy, suggesting that TML is a promising predictive biomarker for immunotherapy. REGISTRATION The study protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO Registration No: CRD42023405596).
Collapse
Affiliation(s)
- Chenghao Ma
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Qiong Teng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Fengying Du
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Gastrointestinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
19
|
Zakariya F, Salem FK, Alamrain AA, Sanker V, Abdelazeem ZG, Hosameldin M, Tan JK, Howard R, Huang H, Awuah WA. Refining mutanome-based individualised immunotherapy of melanoma using artificial intelligence. Eur J Med Res 2024; 29:25. [PMID: 38183141 PMCID: PMC10768232 DOI: 10.1186/s40001-023-01625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024] Open
Abstract
Using the particular nature of melanoma mutanomes to develop medicines that activate the immune system against specific mutations is a game changer in immunotherapy individualisation. It offers a viable solution to the recent rise in resistance to accessible immunotherapy alternatives, with some patients demonstrating innate resistance to these drugs despite past sensitisation to these agents. However, various obstacles stand in the way of this method, most notably the practicality of sequencing each patient's mutanome, selecting immunotherapy targets, and manufacturing specific medications on a large scale. With the robustness and advancement in research techniques, artificial intelligence (AI) is a potential tool that can help refine the mutanome-based immunotherapy for melanoma. Mutanome-based techniques are being employed in the development of immune-stimulating vaccines, improving current options such as adoptive cell treatment, and simplifying immunotherapy responses. Although the use of AI in these approaches is limited by data paucity, cost implications, flaws in AI inference capabilities, and the incapacity of AI to apply data to a broad population, its potential for improving immunotherapy is limitless. Thus, in-depth research on how AI might help the individualisation of immunotherapy utilising knowledge of mutanomes is critical, and this should be at the forefront of melanoma management.
Collapse
Affiliation(s)
- Farida Zakariya
- Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Nigeria
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Fatma K Salem
- Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | | | - Vivek Sanker
- Research Assistant, Dept. Of Neurosurgery, Trivandrum Medical College, Trivandrum, India
| | - Zainab G Abdelazeem
- Division of Molecular Biology, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | | | | | - Rachel Howard
- School of Clinical Medicine, University of Cambridge, Cambridge, England
| | - Helen Huang
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Wireko Andrew Awuah
- Medical Institute, Sumy State University, Zamonstanksya 7, Sumy, 40007, Ukraine.
| |
Collapse
|
20
|
Xue D, Peng H, Li Z, Xu J, Ma H, Dang Y, Li F, Wang G, Sun Q. Comprehensive analysis reveals TSPEAR as a prognostic biomarker in colorectal cancer. J Cancer 2024; 15:809-824. [PMID: 38213725 PMCID: PMC10777046 DOI: 10.7150/jca.90028] [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: 09/11/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most common malignant tumors and has high morbidity and mortality rates. Previous studies have shown that TSPEAR mutations are involved in the development and progression of gastric cancer and liver cancer. However, the role of TSPEAR in CRC is still unclear. Methods: In The Cancer Genome Atlas (TCGA) database, 590 CRC patients with complete survival information were analyzed. We assessed TSPEAR expression in a pan-cancer dataset from the TCGA database. Cox regression analysis was performed to evaluate factors associated with prognosis. Enrichment analysis via the R package "clusterProfiler" was used to explore the potential function of TSPEAR. The single-sample GSEA (ssGSEA) method from the R package "GSVA" and the TIMER database were used to investigate the association between the immune infiltration level and TSPEAR expression in CRC. The R package "maftools" was used to explore the association between tumour mutation burden (TMB) and TSPEAR expression in CRC. CCK-8 assays and cell invasion assays were used to detect the effect of TSPEAR and TGIF2 on the biological behavior of CRC cells. Results: Pan-cancer analysis revealed that TSPEAR was upregulated in CRC tissues compared to normal tissues and that high TSPEAR expression was associated with poorer overall survival (OS) (p=0.0053). The expression of TSPEAR increased with increasing TNM stage, T stage, N stage, and M stage. The nomogram constructed with TSPEAR, age, and TNM stage showed better predictive value than TSPEAR, age, or TNM stage alone. Immune cell infiltration analysis revealed that high expression of TSPEAR was associated with lower immune cell infiltration. Tumor mutation burden (TMB) analysis indicated that high expression of TSPEAR was associated with lower TMB (p=0.005), and high TMB was associated with shorter OS (p=0.02). CCK-8 assays and cell invasion assays indicated that in vitro knockdown of TSPEAR inhibited the proliferation, migration, and invasion of CRC cells. In addition, TSPEAR expression may be regulated by the upstream transcription factor TGIF2. Conclusion: TSPEAR expression was higher in CRC tissues than in normal tissues. Its upregulation was significantly associated with a poor prognosis. Additionally, TSPEAR plays a significant role in tumor immunity and the biological behavior of CRC cells. Thus, TSPEAR may become a promising prognostic biomarker and therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Dong Xue
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hang Peng
- Department of Talent Highland, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenghui Li
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiarui Xu
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haiyun Ma
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yueyan Dang
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fanni Li
- Department of Talent Highland, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guanghui Wang
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
21
|
Ahmed J, Das B, Shin S, Chen A. Challenges and Future Directions in the Management of Tumor Mutational Burden-High (TMB-H) Advanced Solid Malignancies. Cancers (Basel) 2023; 15:5841. [PMID: 38136385 PMCID: PMC10741991 DOI: 10.3390/cancers15245841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
A standardized assessment of Tumor Mutational Burden (TMB) poses challenges across diverse tumor histologies, treatment modalities, and testing platforms, requiring careful consideration to ensure consistency and reproducibility. Despite clinical trials demonstrating favorable responses to immune checkpoint inhibitors (ICIs), not all patients with elevated TMB exhibit benefits, and certain tumors with a normal TMB may respond to ICIs. Therefore, a comprehensive understanding of the intricate interplay between TMB and the tumor microenvironment, as well as genomic features, is crucial to refine its predictive value. Bioinformatics advancements hold potential to improve the precision and cost-effectiveness of TMB assessments, addressing existing challenges. Similarly, integrating TMB with other biomarkers and employing comprehensive, multiomics approaches could further enhance its predictive value. Ongoing collaborative endeavors in research, standardization, and clinical validation are pivotal in harnessing the full potential of TMB as a biomarker in the clinic settings.
Collapse
Affiliation(s)
- Jibran Ahmed
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Sarah Shin
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Alice Chen
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Ke L, Li S, Huang D. The predictive value of tumor mutation burden on survival of gastric cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Int Immunopharmacol 2023; 124:110986. [PMID: 37748223 DOI: 10.1016/j.intimp.2023.110986] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Tumor mutation burden (TMB) is a complement to traditional biomarkers related to the efficacy of immune checkpoint inhibitors (ICIs). The relationship between TMB and the efficacy of ICIs in gastric cancer was controversial. The systematic review and meta-analysis were conducted to investigate the predictive value of TMB on survival of gastric cancer patients treated with ICIs. METHODS We searched the databases PubMed, Embase, and Web of Science for articles, then screened eligible articles according to inclusion criteria. The effective data were extracted to calculate the pooled effects of hazard ratio (HR) for overall survival (OS) and progression-free survival (PFS), then perform publication bias, sensitivity analysis, and subgroup analysis by STATA 16.0. RESULTS The high TMB patients showed significantly longer survival than the low TMB patients (OS: HR 0.65,95% CI 0.55, 0.77, p < 0.001; PFS: HR 0.51, 95% CI 0.33, 0.77, p = 0.001). In the Asian subgroup, patients with high TMB exhibited better prognosis compared to low TMB (OS: HR 0.56, 95% CI 0.43, 0.72, p < 0.001; PFS: HR 0.45, 95% CI 0.28, 0.72, p = 0.001). In the non-Asian subgroup, the survival benefit was observed to be skewed toward patients with high TMB, but it was not statistically significant (OS:HR 0.61, 95% CI 0.32, 1.16, p = 0.133; PFS:HR 0.68, 95% CI 0.31, 1.48, p = 0.322). CONCLUSIONS This meta-analysis demonstrated that gastric cancer patients with high TMB showed significant benefits from ICIs compared to those with low TMB patients, particularly in Asian populations.
Collapse
Affiliation(s)
- Liyuan Ke
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Danxue Huang
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
23
|
Anzar I, Malone B, Samarakoon P, Vardaxis I, Simovski B, Fontenelle H, Meza-Zepeda LA, Stratford R, Keung EZ, Burgess M, Tawbi HA, Myklebost O, Clancy T. The interplay between neoantigens and immune cells in sarcomas treated with checkpoint inhibition. Front Immunol 2023; 14:1226445. [PMID: 37799721 PMCID: PMC10548483 DOI: 10.3389/fimmu.2023.1226445] [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: 05/21/2023] [Accepted: 07/10/2023] [Indexed: 10/07/2023] Open
Abstract
Introduction Sarcomas are comprised of diverse bone and connective tissue tumors with few effective therapeutic options for locally advanced unresectable and/or metastatic disease. Recent advances in immunotherapy, in particular immune checkpoint inhibition (ICI), have shown promising outcomes in several cancer indications. Unfortunately, ICI therapy has provided only modest clinical responses and seems moderately effective in a subset of the diverse subtypes. Methods To explore the immune parameters governing ICI therapy resistance or immune escape, we performed whole exome sequencing (WES) on tumors and their matched normal blood, in addition to RNA-seq from tumors of 31 sarcoma patients treated with pembrolizumab. We used advanced computational methods to investigate key immune properties, such as neoantigens and immune cell composition in the tumor microenvironment (TME). Results A multifactorial analysis suggested that expression of high quality neoantigens in the context of specific immune cells in the TME are key prognostic markers of progression-free survival (PFS). The presence of several types of immune cells, including T cells, B cells and macrophages, in the TME were associated with improved PFS. Importantly, we also found the presence of both CD8+ T cells and neoantigens together was associated with improved survival compared to the presence of CD8+ T cells or neoantigens alone. Interestingly, this trend was not identified with the combined presence of CD8+ T cells and TMB; suggesting that a combined CD8+ T cell and neoantigen effect on PFS was important. Discussion The outcome of this study may inform future trials that may lead to improved outcomes for sarcoma patients treated with ICI.
Collapse
Affiliation(s)
- Irantzu Anzar
- Oslo Cancer Cluster, NEC OncoImmunity AS, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | - Leonardo A. Meza-Zepeda
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Core Facilities, Oslo University Hospital, Oslo, Norway
| | | | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Melissa Burgess
- Department of Medical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ola Myklebost
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Trevor Clancy
- Oslo Cancer Cluster, NEC OncoImmunity AS, Oslo, Norway
| |
Collapse
|
24
|
Parent P, Marcq G, Adeleke S, Turpin A, Boussios S, Rassy E, Penel N. Predictive biomarkers for immune checkpoint inhibitor response in urothelial cancer. Ther Adv Med Oncol 2023; 15:17588359231192402. [PMID: 37692364 PMCID: PMC10486227 DOI: 10.1177/17588359231192402] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/19/2023] [Indexed: 09/12/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are commonly used to treat patients with advanced urothelial cancer. However, a significant number of patients do not respond to ICI, and the lack of validated predictive biomarkers impedes the success of the ICI strategy alone or in combination with chemotherapy or targeted therapies. In addition, some patients experience potentially severe adverse events with limited clinical benefit. Therefore, identifying biomarkers of response to ICI is crucial to guide treatment decisions. The most evaluated biomarkers to date are programmed death ligand 1 expression, microsatellite instability/defective mismatch repair phenotype, and tumor mutational burden. Other emerging biomarkers, such as circulating tumor DNA and microbiota, require evaluation in clinical trials. This review aims to examine these biomarkers for ICI response in urothelial cancer and assess their analytical and clinical validation.
Collapse
Affiliation(s)
- Pauline Parent
- Medical Oncology Department, Centre Hospitalier Universitaire de Lille (CHU Lille), University of Lille, Hôpital Huriez, Lille 59037, France
| | - Gautier Marcq
- Urology Department, Claude Huriez Hospital, Centre Hospitalier Universitaire de Lille (CHU Lille), Lille, France
| | - Sola Adeleke
- Department of Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- King’s College London, Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, London, UK
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020U1277 – CANTHER – CANcer Heterogeneity Plasticity and Resistance to THERapies, University of Lille, Lille, France
| | - Anthony Turpin
- Medical Oncology Department, CHU Lille, University of Lille, Lille, France
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 – UMR-S 1277 – CANTHER – CANcer Heterogeneity, Plasticity and Resistance to THERapies, University of Lille, Lille, France
| | - Stergios Boussios
- Department of Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Medway NHS Foundation Trust, Gillingham, Kent, UK
- AELIA Organization, Thessaloniki, Greece
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, Villejuif, France
| | - Nicolas Penel
- Centre Oscar Lambret, Clinical Research and Innovation Department, Medical Oncology Department, Lille, France
- University of Lille, CHU Lille, ULR 2694 – Metrics: Evaluation des technologies de santé et des pratiques médicales, Lille, France
| |
Collapse
|
25
|
Scobie MR, Zhou KI, Ahmed S, Kelley MJ. Utility of Tumor Mutational Burden as a Biomarker for Response to Immune Checkpoint Inhibition in the VA Population. JCO Precis Oncol 2023; 7:e2300176. [PMID: 38039430 DOI: 10.1200/po.23.00176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 12/03/2023] Open
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) are used for an increasing number of indications across various tumor types, as well as several tumor-agnostic indications in patients with advanced cancer. Although many patients benefit from ICI therapy, others do not, highlighting a need for better predictive biomarkers. Tumor mutational burden (TMB) reflects the global number of mutations within a tumor and has been widely explored as a predictive biomarker of ICI response. The current tumor type-agnostic US Food and Drug Administration approval of pembrolizumab for metastatic solid tumors defines high TMB (TMB-H) as ≥10 mut/Mb as measured by FoundationOne CDx. This fixed cutoff may not be the ideal value across all solid tumors. METHODS We performed a retrospective analysis of the association of survival outcomes with TMB in patients treated with ICI for five major cancer types, using real-world data from the VA. Survival was measured from initiation of ICI, and Kaplan-Meier survival curves were compared by log-rank test. RESULTS Overall survival (OS) was significantly longer for patients with TMB-H versus TMB low tumors in non-small-cell lung cancer (NSCLC; n = 1,593), head and neck (H&N) cancer (n = 222), and urothelial cancer (n = 332). OS was not significantly different based on TMB status in melanoma (n = 207) or esophageal/gastric cancer (n = 248). CONCLUSION Consistent with previous studies, a predictive value of TMB ≥10 mut/Mb for ICI response was found in NSCLC and H&N, but not in esophageal/gastric cancer. Although inconclusive in the literature, significant association was found in urothelial cancer. The predictive value of TMB in melanoma was inconclusive. Our analysis does not support the use of a fixed threshold for TMB as a standalone predictive biomarker for ICI across all solid tumors.
Collapse
Affiliation(s)
- Micaela R Scobie
- Department of Veterans Affairs, National Oncology Program, Washington, DC
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
| | - Katherine I Zhou
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC
| | - Sara Ahmed
- Department of Veterans Affairs, National Oncology Program, Washington, DC
| | - Michael J Kelley
- Department of Veterans Affairs, National Oncology Program, Washington, DC
- Division of Hematology-Oncology, Durham VA Medical Center, Durham, NC
| |
Collapse
|
26
|
Wankhede D, Grover S, Hofman P. The prognostic value of TMB in early-stage non-small cell lung cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2023; 15:17588359231195199. [PMID: 37667779 PMCID: PMC10475237 DOI: 10.1177/17588359231195199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
Background Tumor mutation burden (TMB) has been validated as a predictive biomarker for immunotherapy response and survival in numerous cancer types. Limited data is available on the inherent prognostic role of TMB in early-stage tumors. Objective To evaluate the prognostic role of TMB in early-stage, resected non-small cell lung cancer (NSCLC). Design Systematic review and meta-analysis of pertinent prospective and retrospective studies. Data sources and methods Publication search was performed in PubMed, Embase, Cochrane Library, and Web of Science databases. Based on the level of heterogeneity, a random- or fixed-effects model was used to calculate pooled effects of hazard ratio (HR) for overall survival (OS) and disease-free survival (DFS). The source of heterogeneity was investigated using sensitivity analysis, subgroup analysis, and publication bias assessment. Results Ten studies comprising 2520 patients were included in this analysis. There was no statistically significant difference in OS (HR, 1.18, 95% CI, 0.70, 1.33; p 0.53, I2 = 80%; phet < 0.0001) and DFS (HR, 1.18, 95% CI, 0.91, 1.52; p = 0.53, I2 = 75%; phet = 0.0001) between the high-TMB and low-TMB group. Subgroup analyses indicated that East Asian ethnicity, and TMB detected using whole exome sequencing, and studies with <100 patients had poor DFS in the high-TMB group. Conclusion The inherent prognostic role of TMB is limited in early-stage NSCLC. Ethnic differences in mutation burden must be considered while designing future trials on neoadjuvant immunotherapy. Further research in the harmonization and standardization of panel-based TMB is essential for its widespread clinical utility.Registration: CRD42023392846.
Collapse
Affiliation(s)
- Durgesh Wankhede
- German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg – Standort Marburg, Marburg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, University Côte d’Azur, Nice, France
- Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081 and UMR CNRS 7284, Team 4, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Pasteur Hospital, Nice, France
- University Hospital Institute RespirERA, Nice, France
- University Hospital Federation OncoAge, CHU de Nice, University Côte d’Azur, Nice, France
| |
Collapse
|
27
|
Chang JW, Huang C, Huang W, Wang Y, Hsieh J, Chang Y, Huang Y, Wu C, Wang Y, Chen S, Tan KT, Chen C, Wu C. Genomic and tumour microenvironmental biomarkers of immune checkpoint inhibitor response in advanced Taiwanese melanoma. Clin Transl Immunology 2023; 12:e1465. [PMID: 37649975 PMCID: PMC10463562 DOI: 10.1002/cti2.1465] [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: 03/25/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023] Open
Abstract
Objective Genomic biomarkers predicting immune checkpoint inhibitor (ICI) treatment outcomes for Asian metastatic melanoma have been rarely reported. This study presents data on next-generation sequencing (NGS) and tumour microenvironment biomarkers in 33 cases. Methods Thirty-three patients with advanced melanoma, who underwent ICI treatment at the Chang Gung Memorial Hospital in Taiwan, were recruited. The study evaluated clinical outcomes, including response rate, disease control rate, progression-free survival (PFS) rate and overall survival (OS) rate. Archived tissue samples from 33 cases were subjected to NGS by ACTOnco, and ACTTME was employed in 25 cases. Results The most prevalent driver mutations were BRAF mutations (24.2%), followed by NRAS (15.2%), KIT (12.1%), KRAS (9.1%) and NF1 (9.1%) mutations. Acral/mucosal melanomas exhibited distinct mutation patterns compared to non-acral melanomas. Tumour mutational burden estimated using ACTOnco was not associated with ICI efficacy. Notably, genetic alterations in the p53 pathway (CDKNA2 loss, MDM2 gain/amplification and TP53 mutation) accounted for 36.4% and were significantly associated with unfavourable PFS (median PFS 2.7 months vs. 3.9 months, P = 0.0394). Moreover, 26 genes were identified as differentially expressed genes that were upregulated in patients with clinical benefits compared to those without benefits. Four genes, GZMH, GZMK, AIM2 and CTLA4, were found to be associated with both PFS and OS. Conclusion Genetic alterations in the p53 pathway may be critical in Asian patients with melanoma undergoing ICI treatment. Further investigation is required to explore this mechanism and validate these findings.
Collapse
Affiliation(s)
- John Wen‐Cheng Chang
- Division of Hematology‐Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chien‐Jung Huang
- Institute of Biomedical InformaticsNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Wen‐Kuan Huang
- Division of Hematology‐Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Yu‐Chao Wang
- Institute of Biomedical InformaticsNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jia‐Juan Hsieh
- Division of Hematology‐Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Yao‐Yu Chang
- Department of Dermatology, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Yen‐Lin Huang
- School of MedicineNational Tsing‐Hua UniversityHsinchuTaiwan
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at LinkouInstitute of Stem Cell and Translational Cancer ResearchTaoyuanTaiwan
| | | | | | | | - Kien Thiam Tan
- ACT Genomics Co., LtdTaipeiTaiwan
- Anbogen Therapeutics Co., LtdTaipeiTaiwan
| | - Chiao‐Ping Chen
- Division of Hematology‐Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chiao‐En Wu
- Division of Hematology‐Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of MedicineChang Gung UniversityTaoyuanTaiwan
| |
Collapse
|
28
|
Gershtein ES, Mochalova AS, Korotkova EA, Samoilova EV, Vashketova OI, Kuz'min YB, Sokolov NY, Kushlinskii NE. Dynamics of Soluble Forms of the Immune Checkpoint Components PD-1/PD-L1/B7-H3, CD314/ULBP1, and HLA-G in Peripheral Blood of Melanoma Patients Receiving Blockers of Programmed Cell Death Protein PD-1. Bull Exp Biol Med 2023; 175:481-486. [PMID: 37773572 DOI: 10.1007/s10517-023-05891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Indexed: 10/01/2023]
Abstract
The content of the soluble forms of immune checkpoint components sPD-1, sPD-L1 in blood serum, and sB7-H3, sCD314, sULBP1, sHLA-G in blood plasma of 30 melanoma patients receiving immunotherapy with anti-PD-1 antibodies (nivolumab, pembrolisumab) was measured before and in 4 and 8 weeks after the start of immunotherapy. The control group comprised 70 practically healthy donors. Standard immunoassay kits were used. In melanoma patients, the levels of sPD-L1 and sB7-H3 were significantly higher than in the control group (p<0001), sPD-1 level did not differ from the control, while sCD314 and sHLA-G levels were insignificantly decreased. During therapy, opposite changes in the levels of markers in individual patients were observed, and frequently after the initial increase (or decrease) after the first 4 weeks normalization did occur in the further 4 weeks. No statistically significant associations between the initial levels of markers and direction of their changes during treatment were found, but some trends indicating to the potential benefits from assessment of soluble forms of immune checkpoint proteins for evaluation and monitoring of the efficiency of the therapy with immune checkpoint blockers were revealed: significant decrease of sB7-H3 and sPD-1 levels in the course of treatment, higher initial sPD-1 level in patients with future progression than in those with stabilization or partial effect, and lower progression frequency in patients with increasing sPD-1 and sPD-L1 levels than in those with decreasing markers levels.
Collapse
Affiliation(s)
- E S Gershtein
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A S Mochalova
- AO GK MEDSI, MEDSI Clinical Hospital in Otradnoe, Krasnogorsk, Russia
| | - E A Korotkova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E V Samoilova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - O I Vashketova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yu B Kuz'min
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N Yu Sokolov
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N E Kushlinskii
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
29
|
Chatziioannou E, Roßner J, Aung TN, Rimm DL, Niessner H, Keim U, Serna-Higuita LM, Bonzheim I, Kuhn Cuellar L, Westphal D, Steininger J, Meier F, Pop OT, Forchhammer S, Flatz L, Eigentler T, Garbe C, Röcken M, Amaral T, Sinnberg T. Deep learning-based scoring of tumour-infiltrating lymphocytes is prognostic in primary melanoma and predictive to PD-1 checkpoint inhibition in melanoma metastases. EBioMedicine 2023; 93:104644. [PMID: 37295047 PMCID: PMC10363450 DOI: 10.1016/j.ebiom.2023.104644] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Recent advances in digital pathology have enabled accurate and standardised enumeration of tumour-infiltrating lymphocytes (TILs). Here, we aim to evaluate TILs as a percentage electronic TIL score (eTILs) and investigate its prognostic and predictive relevance in cutaneous melanoma. METHODS We included stage I to IV cutaneous melanoma patients and used hematoxylin-eosin-stained slides for TIL analysis. We assessed eTILs as a continuous and categorical variable using the published cut-off of 16.6% and applied Cox regression models to evaluate associations of eTILs with relapse-free, distant metastasis-free, and overall survival. We compared eTILs of the primaries with matched metastasis. Moreover, we assessed the predictive relevance of eTILs in therapy-naïve metastases according to the first-line therapy. FINDINGS We analysed 321 primary cutaneous melanomas and 191 metastatic samples. In simple Cox regression, tumour thickness (p < 0.0001), presence of ulceration (p = 0.0001) and eTILs ≤16.6% (p = 0.0012) were found to be significant unfavourable prognostic factors for RFS. In multiple Cox regression, eTILs ≤16.6% (p = 0.0161) remained significant and downgraded the current staging. Lower eTILs in the primary tissue was associated with unfavourable relapse-free (p = 0.0014) and distant metastasis-free survival (p = 0.0056). In multiple Cox regression adjusted for tumour thickness and ulceration, eTILs as continuous remained significant (p = 0.019). When comparing TILs in primary tissue and corresponding metastasis of the same patient, eTILs in metastases was lower than in primary melanomas (p < 0.0001). In therapy-naïve metastases, an eTILs >12.2% was associated with longer progression-free survival (p = 0.037) and melanoma-specific survival (p = 0.0038) in patients treated with anti-PD-1-based immunotherapy. In multiple Cox regression, lactate dehydrogenase (p < 0.0001) and eTILs ≤12.2% (p = 0.0130) were significantly associated with unfavourable melanoma-specific survival. INTERPRETATION Assessment of TILs is prognostic in primary melanoma samples, and the eTILs complements staging. In therapy-naïve metastases, eTILs ≤12.2% is predictive of unfavourable survival outcomes in patients receiving anti-PD-1-based therapy. FUNDING See a detailed list of funding bodies in the Acknowledgements section at the end of the manuscript.
Collapse
Affiliation(s)
- Eftychia Chatziioannou
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Jana Roßner
- Department of Dermatology, University of Heidelberg, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany
| | - Thazin New Aung
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Heike Niessner
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Ulrike Keim
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| | - Lina Maria Serna-Higuita
- Department of Clinical Epidemiology and Applied Biostatistics, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Luis Kuhn Cuellar
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Dana Westphal
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Skin Cancer Center at the University Cancer Center and National Center for Tumor Diseases, Technical University Dresden, 01307 Dresden, Germany
| | - Julian Steininger
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Skin Cancer Center at the University Cancer Center and National Center for Tumor Diseases, Technical University Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Skin Cancer Center at the University Cancer Center and National Center for Tumor Diseases, Technical University Dresden, 01307 Dresden, Germany
| | - Oltin Tiberiu Pop
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Stephan Forchhammer
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Thomas Eigentler
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Claus Garbe
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany
| | - Tobias Sinnberg
- Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Tübingen, Germany; Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
30
|
Daunke T, Beckinger S, Rahn S, Krüger S, Heckl S, Schäfer H, Wesch D, Pilarsky C, Eckstein M, Hartmann A, Röcken C, Wandmacher AM, Sebens S. Expression and role of the immune checkpoint regulator PD-L1 in the tumor-stroma interplay of pancreatic ductal adenocarcinoma. Front Immunol 2023; 14:1157397. [PMID: 37449210 PMCID: PMC10337136 DOI: 10.3389/fimmu.2023.1157397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Immune checkpoint inhibitors (ICI), e.g., targeting programmed cell death protein 1-ligand 1 (PD-L1) or its receptor PD-1, have markedly improved the therapy of many cancers but so far failed in pancreatic ductal adenocarcinoma (PDAC). Macrophages represent one of the most abundant immune cell populations within the tumor microenvironment (TME) of PDAC being able to either support or restrain tumor progression depending on their phenotype. To better understand treatment failure of PD-L1/PD-1 inhibitors in PDAC, this study examined PD-L1 expression in the context of a dynamic TME in PDAC with a particular focus on the impact of macrophages. Methods Formalin-fixed and paraffin embedded tissue samples of primary PDAC tissues and corresponding liver metastases were used for immunohistochemical analyses. Serial sections were stained with antibodies detecting Pan-Cytokeratin, CD68, CD163, CD8, and PD-L1.To investigate whether the PD-1/PD-L1 axis and macrophages contribute to immune escape of PDAC cells, a stroma enriched 3D spheroid coculture model was established in vitro, using different PDAC cell lines and macrophages subtypes as well as CD8+ T cells. Functional and flow cytometry analyses were conducted to characterize cell populations. Results Immunohistochemical analyses revealed that PD-L1 is mainly expressed by stroma cells, including macrophages and not PDAC cells in primary PDAC tissues and corresponding liver metastases. Notably, high local abundance of macrophages and strong PD-L1 staining were commonly found at invasion fronts of tumoral lesions between CD8+ T cells and tumor cells. In order to investigate whether PD-L1 expressing macrophages impact the response of PDAC cells to treatment with PD-L1/PD-1 inhibitors, we developed a spheroid model comprising two different PDAC cell lines and different ratios of in vitro differentiated primary M1- or M2-like polarized macrophages. In line with our in situ findings, high PD-L1 expression was observed in macrophages rather than PDAC cells, which was further increased by the presence of PDAC cells. The effector phenotype of co-cultured CD8+ T cells exemplified by expression of activation markers and release of effector molecules was rather enhanced by PDAC macrophage spheroids, particularly with M1-like macrophages compared to mono-culture spheroids. However, this was not associated with enhanced PDAC cell death. ICI treatment with either Durvalumab or Pembrolizumab alone or in combination with Gemcitabine hardly affected the effector phenotype of CD8+ T cells along with PDAC cell death. Thus, despite strong PD-L1 expression in macrophages, ICI treatment did not result in an enhanced activation and cytotoxic phenotype of CD8+ T cells. Conclusion Overall, our study revealed novel insights into the interplay of PDAC cells and macrophages in the presence of ICI.
Collapse
Affiliation(s)
- Tina Daunke
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Silje Beckinger
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sascha Rahn
- Biochemical Institute, Kiel University, Kiel, Germany
| | - Sandra Krüger
- Institute of Pathology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Steffen Heckl
- Institute of Pathology, University Medical Center Schleswig-Holstein, Kiel, Germany
- Department of Internal Medicine II, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Heiner Schäfer
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, Kiel University and University Medical Center Schleswig-Holstein (UKSH), Kiel, Germany
| | - Christian Pilarsky
- Translational Research Center, University Hospital Erlangen, Erlangen, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christoph Röcken
- Institute of Pathology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Anna Maxi Wandmacher
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Internal Medicine II, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
31
|
Bushara O, Tidwell J, Wester JR, Miura J. The Current State of Neoadjuvant Therapy in Resectable Advanced Stage Melanoma. Cancers (Basel) 2023; 15:3344. [PMID: 37444454 DOI: 10.3390/cancers15133344] [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: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The advent of effective immunotherapy and targeted therapy has significantly improved outcomes in advanced-stage resectable melanoma. Currently, the mainstay of treatment of malignant melanoma is surgery followed by adjuvant systemic therapies. However, recent studies have shown a potential role for neoadjuvant therapy in the treatment of advanced-stage resectable melanoma. Mechanistically, neoadjuvant immunotherapy may yield a more robust response than adjuvant immunotherapy, as the primary tumor serves as an antigen in this setting rather than only micrometastatic disease after the index procedure. Additionally, targeted therapy has been shown to yield effective neoadjuvant cytoreduction, and oncolytic viruses may also increase the immunogenicity of primary tumors. Effective neoadjuvant therapy may serve to decrease tumor size and thus reduce the extent of required surgery and thus morbidity. It also allows for assessment of pathologic response, facilitating prognostication as well as tailoring future therapy. The current literature consistently supports that neoadjuvant therapy, even as little as one dose, is associated with improved outcomes and is well-tolerated. Some patients with a complete pathological response may even avoid surgery completely. These results challenge the current paradigm of a surgery-first approach and provide further evidence supporting neoadjuvant therapy in advanced-stage resectable melanoma. Further research into the optimal treatment schedule and dose timing is warranted, as is the continued investigation of novel therapies and combinations of therapies.
Collapse
Affiliation(s)
- Omar Bushara
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerica Tidwell
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James R Wester
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Miura
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Li LL, Yu CF, Xie HT, Chen Z, Jia BH, Xie FY, Cai YF, Xue P, Zhu SJ. Biomarkers and factors in small cell lung cancer patients treated with immune checkpoint inhibitors: A meta-analysis. Cancer Med 2023. [PMID: 37161541 DOI: 10.1002/cam4.5800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/18/2023] [Accepted: 02/25/2023] [Indexed: 05/11/2023] Open
Abstract
OBJECTIVE The aim of this meta-analysis was to summarize the available results of immunotherapy predictors for small cell lung cancer (SCLC) and to provide evidence-based information for their potential predictive value of efficacy. METHODS We searched PubMed, EMBASE, Web of Science, The Cochrane Library, and ClinicalTrials (from January 1, 1975 to November 1, 2021). The hazard ratios (HR) and its 95% confidence intervals (CIs) and tumor response rate of the included studies were extracted. RESULTS Eleven studies were eventually included and the pooled results showed that programmed cell death ligand 1 (PD-L1) positive: objective response rate (ORR) (relative risk [RR] = 1.39, 95% CI [0.48, 4.03], p = 0.54), with high heterogeneity (p = 0.05, I2 = 56%); disease control rate [DCR] (RR = 1.31, 95% CI [0.04, 38.57], p = 0.88), with high heterogeneity (p = 0.04, I2 = 75%); overall survival (OS) (HR = 0.89, 95% CI [0.74, 1.07], p = 0.22); and progression-free survival (PFS) (HR = 0.83, 95% CI [0.59, 1.16], p = 0.27), with high heterogeneity (p = 0.005, I2 = 73.1%). TMB-High (TMB-H): OS (HR = 0.86, 95% CI [0.74, 1.00], p = 0.05); PFS (HR = 0.71, 95% CI [0.6, 0.85], p < 0.001). Lactate dehydrogenase (LDH) >upper limit of normal (ULN): OS (HR = 0.95, 95% CI [0.81, 1.11], p = 0.511). Asian patients: OS (HR = 0.87, 95% CI [0.72, 1.04], p = 0.135); White/Non-Asian patients: OS (HR = 0.83, 95% CI [0.76, 0.90], p < 0.001). Liver metastasis patients: OS (HR = 0.93, 95% CI [0.83, 1.05], p = 0.229); PFS (HR = 0.84, 95% CI [0.67, 1.06], p = 0.141). Central nervous system (CNS) metastasis patients: OS (HR = 0.91, 95% CI [0.71, 1.17], p = 0.474); PFS (HR = 1.03, 95% CI [0.66, 1.60], p = 0.903). CONCLUSION The available research results do not support the recommendation of PD-L1 positive and TMB-H as predictors for the application of immune checkpoint inhibitors (ICIs) in SCLC patients. LDH, baseline liver metastasis and CNS metastasis may be used as markers/influencing factors for predicting the efficacy of ICIs in SCLC patients. Non-Asian SCLC patients had better efficacy with ICIs in our results.
Collapse
Affiliation(s)
- Lin-Lu Li
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Cheng-Feng Yu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| | - Hong-Ting Xie
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Zheng Chen
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Bo-Hui Jia
- Beijing Sihui West District Hospital, 100082, Beijing, China
| | - Fei-Yu Xie
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Ya-Fang Cai
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Peng Xue
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| | - Shi-Jie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, 100102, Beijing, China
| |
Collapse
|
33
|
Eroglu Z, Krinshpun S, Kalashnikova E, Sudhaman S, Ozturk Topcu T, Nichols M, Martin J, Bui KM, Palsuledesai CC, Malhotra M, Olshan P, Markowitz J, Khushalani NI, Tarhini AA, Messina JL, Aleshin A. Circulating tumor DNA-based molecular residual disease detection for treatment monitoring in advanced melanoma patients. Cancer 2023; 129:1723-1734. [PMID: 36869646 DOI: 10.1002/cncr.34716] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 03/05/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have substantially improved overall survival in patients with advanced melanoma; however, the lack of biomarkers to monitor treatment response and relapse remains an important clinical challenge. Thus, a reliable biomarker is needed that can risk-stratify patients for disease recurrence and predict response to treatment. METHODS A retrospective analysis using a personalized, tumor-informed circulating tumor DNA (ctDNA) assay on prospectively collected plasma samples (n = 555) from 69 patients with advanced melanoma was performed. Patients were divided into three cohorts: cohort A (N = 30), stage III patients receiving adjuvant ICI/observation; cohort B (N = 29), unresectable stage III/IV patients receiving ICI therapy; and cohort C (N = 10), stage III/IV patients on surveillance after planned completion of ICI therapy for metastatic disease. RESULTS In cohort A, compared to molecular residual disease (MRD)-negative patients, MRD-positivity was associated with significantly shorter distant metastasis-free survival (DMFS; hazard ratio [HR], 10.77; p = .01). Increasing ctDNA levels from the post-surgical or pre-treatment time point to after 6 weeks of ICI were predictive of shorter DMFS in cohort A (HR, 34.54; p < .0001) and shorter progression-free survival (PFS) in cohort B (HR, 22; p = .006). In cohort C, all ctDNA-negative patients remained progression-free for a median follow-up of 14.67 months, whereas ctDNA-positive patients experienced disease progression. CONCLUSION Personalized and tumor-informed longitudinal ctDNA monitoring is a valuable prognostic and predictive tool that may be used throughout the clinical course of patients with advanced melanoma.
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | | | | | - Turkan Ozturk Topcu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Matt Nichols
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Justin Martin
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Katherine M Bui
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | | | | | - Joseph Markowitz
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Nikhil I Khushalani
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Ahmad A Tarhini
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Jane L Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | |
Collapse
|
34
|
Identification of Pyroptosis-Relevant Signature in Tumor Immune Microenvironment and Prognosis in Skin Cutaneous Melanoma Using Network Analysis. Stem Cells Int 2023; 2023:3827999. [PMID: 36818162 PMCID: PMC9931490 DOI: 10.1155/2023/3827999] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/19/2022] [Accepted: 11/25/2022] [Indexed: 02/10/2023] Open
Abstract
Background Pyroptosis is closely related to the programmed death of cancer cells as well as the tumor immune microenvironment (TIME) via the host-tumor crosstalk. However, the role of pyroptosis-related genes as prognosis and TIME-related biomarkers in skin cutaneous melanoma (SKCM) patients remains unknown. Methods We evaluated the expression profiles, copy number variations, and somatic mutations (CNVs) of 27 genes obtained from MSigDB database regulating pyroptosis among TCGA-SKCM patients. Thereafter, we conducted single-sample gene set enrichment analysis (ssGSEA) for evaluating pyroptosis-associated expression patterns among cases and for exploring the associations with clinicopathological factors and prognostic outcome. In addition, a prognostic pyroptosis-related signature (PPRS) model was constructed by performing Cox regression, weighted gene coexpression network analysis (WGCNA), and least absolute shrinkage and selection operator (LASSO) analysis to score SKCM patients. On the other hand, we plotted the ROC and survival curves for model evaluation and verified the robustness of the model through external test sets (GSE22153, GSE54467, and GSE65904). Meanwhile, we examined the relations of clinical characteristics, oncogene mutations, biological processes (BPs), tumor stemness, immune infiltration degrees, immune checkpoints (ICs), and treatment response with PPRS via multiple methods, including immunophenoscore (IPS) analysis, gene set variation analysis (GSVA), ESTIMATE, and CIBERSORT. Finally, we constructed a nomogram incorporating PPRS and clinical characteristics to improve risk evaluation of SKCM. Results Many pyroptosis-regulated genes showed abnormal expression within SKCM. TP53, TP63, IL1B, IL18, IRF2, CASP5, CHMP4C, CHMP7, CASP1, and GSDME were detected with somatic mutations, among which, a majority displayed CNVs at high frequencies. Pyroptosis-associated profiles established based on pyroptosis-regulated genes showed markedly negative relation to low stage and superior prognostic outcome. Blue module was found to be highly positively correlated with pyroptosis. Later, this study established PPRS based on the expression of 8 PAGs (namely, GBP2, HPDL, FCGR2A, IFITM1, HAPLN3, CCL8, TRIM34, and GRIPAP1), which was highly associated with OS, oncogene mutations, tumor stemness, immune infiltration degrees, IC levels, treatment responses, and multiple biological processes (including cell cycle and immunoinflammatory response) in training and test set samples. Conclusions Based on our observations, analyzing modification patterns associated with pyroptosis among diverse cancer samples via PPRS is important, which can provide more insights into TIME infiltration features and facilitate immunotherapeutic development as well as prognosis prediction.
Collapse
|
35
|
Liu M, Li W, Ma X, Che Y, Wei B, Chen M, Zhong L, Zhao S, Chen A, Pang Y, Zeng J, Guo J. Gradient differences of immunotherapy efficacy in metastatic melanoma related to sunlight exposure pattern: A population-based study. Front Oncol 2023; 12:1086664. [PMID: 36686834 PMCID: PMC9850161 DOI: 10.3389/fonc.2022.1086664] [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: 11/01/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have revolutionized metastatic melanoma (MM) treatment in just a few years. Ultraviolet (UV) in sunlight is the most significant environmental cause of melanoma, which is considered to be the main reason for tumor mutation burden (TMB) increase in melanoma. High TMB usually predicts that PD-1 inhibitors are effective. The sunlight exposure pattern of MM might be a clinical feature that matches TMB. The relationship between sunlight exposure patterns and immunotherapy response in MM is unclear. This study aims to investigate the correlation between sunlight exposure patterns and immunotherapy response in MM and establish nomograms that predict 3- and 5-year overall survival (OS) rate. Methods We searched the Surveillance, Epidemiology, and End Results (SEER) database and enrolled MM cases from 2005-2016. According to the advent of ICIs in 2011, the era was divided into the non-ICIs era (2005-2010) and the ICIs era (2011-2016). Patients were divided into three cohorts according to the primary site sunlight exposure patterns: head and neck in the first cohort, trunk arms and legs in the second cohort, and acral sites in the third cohort. We compared survival differences for each cohort between the two eras, performed stratified analysis, established nomograms for predicting 3- and 5-year OS rate, and performed internal validation. Results Comparing the survival difference between the ICIs and non-ICIs era, head and neck melanoma showed the greatest improvement in survival, with 3- and 5-year OS rate increasing by 10.2% and 9.1%, respectively (P=0.00011). In trunk arms and legs melanoma, the 3- and 5-year OS rate increased by 4.6% and 3.9%, respectively (P<0.0001). There is no improvement in survival in acral melanoma (AM) between the two eras (P=0.78). The receiver operating characteristic (ROC) curve, area under the ROC curve (AUC) and calibration graphs show good discrimination and accuracy of nomograms. Decision curve analysis (DCA) suggests good clinical utility of nomograms. Conclusions Based on the classification of sunlight exposure patterns, there is a gradient difference in immunotherapy efficacy for MM. The degree of sunlight exposure is positively correlated with immunotherapy response. The nomograms are sufficiently accurate to predict 3- and 5-year OS rate for MM, allowing for individualized clinical decisions for future clinical work.
Collapse
Affiliation(s)
- Mengsong Liu
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenyuan Li
- Sichuan Evidence-Based Medicine Center of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhui Che
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Wei
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mulan Chen
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Zhong
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siqi Zhao
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anjing Chen
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaobin Pang
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Guo
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Ke L, Li S, Cui H. The prognostic role of tumor mutation burden on survival of breast cancer: a systematic review and meta-analysis. BMC Cancer 2022; 22:1185. [PMID: 36397030 PMCID: PMC9673350 DOI: 10.1186/s12885-022-10284-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background As a potential genetic biomarker, tumor mutation burden (TMB) has made progress in numerous tumors. There are limited data regarding TMB and its prognostic role is controversial in breast cancer. This systematic review and meta-analysis were conducted to assess the prognostic value of TMB on survival of breast cancer. Methods The databases PubMed, Embase, Web of Science, and Cochrane Library were searched for articles published through May 31, 2022. Moreover, effective data were extracted from included studies and calculated pooled effects of hazard ratio (HR) for overall survival (OS) and progression-free survival (PFS) by STATA 16.0. Heterogeneity was conducted by the I2 statistic and p-value. Using publication bias evaluation, sensitivity analysis, and subgroup analysis, the origin of heterogeneity was further investigated. Results They were up to 1,722 patients collected from sixteen cohorts for this analysis. The pooled effects of HR for both OS (HR: 1.14, 95% CI: 0.83,1.58, p > 0.01) and PFS (HR: 0.96, 95% CI: 0.53,1.71, p > 0.01) indicated no statistically significant difference in the high TMB and low TMB group. In immune checkpoint inhibitors (ICIs) subgroup, high TMB patients demonstrated benefit of OS (HR: 0.72, 95% CI: 0.59,0.87, p = 0.001) and PFS (HR: 0.52, 95% CI: 0.35,0.77, p < 0.001), whereas difference was not statistically significant in the non-ICIs subgroup (OS, HR:1.76, 95% CI: 0.97,3.20, p = 0.062; PFS, HR:2.31, 95% CI: 0.89,5.97, p = 0.086). In addition, sensitivity analysis revealed that the pooled effects were stable. The funnel plot and Begg's test suggested the absence of publication bias. Conclusion Meta-analysis revealed that the prognostic relevance of TMB in breast cancer is limited in scope. High TMB may be associated with longer survival only in ICIs-based treatment, but the association is not evident in non-ICIs-based treatment. Trial registration [https://www.crd.york.ac.uk/PROSPERO], Prospective Register of Systematic Reviews (PROSPERO), identifier: CRD42022342488. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10284-1.
Collapse
|
37
|
Hossain SM, Gimenez G, Stockwell PA, Tsai P, Print CG, Rys J, Cybulska-Stopa B, Ratajska M, Harazin-Lechowska A, Almomani S, Jackson C, Chatterjee A, Eccles MR. Innate immune checkpoint inhibitor resistance is associated with melanoma sub-types exhibiting invasive and de-differentiated gene expression signatures. Front Immunol 2022; 13:955063. [PMID: 36248850 PMCID: PMC9554309 DOI: 10.3389/fimmu.2022.955063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Melanoma is a highly aggressive skin cancer, which, although highly immunogenic, frequently escapes the body’s immune defences. Immune checkpoint inhibitors (ICI), such as anti-PD1, anti-PDL1, and anti-CTLA4 antibodies lead to reactivation of immune pathways, promoting rejection of melanoma. However, the benefits of ICI therapy remain limited to a relatively small proportion of patients who do not exhibit ICI resistance. Moreover, the precise mechanisms underlying innate and acquired ICI resistance remain unclear. Here, we have investigated differences in melanoma tissues in responder and non-responder patients to anti-PD1 therapy in terms of tumour and immune cell gene-associated signatures. We performed multi-omics investigations on melanoma tumour tissues, which were collected from patients before starting treatment with anti-PD1 immune checkpoint inhibitors. Patients were subsequently categorized into responders and non-responders to anti-PD1 therapy based on RECIST criteria. Multi-omics analyses included RNA-Seq and NanoString analysis. From RNA-Seq data we carried out HLA phenotyping as well as gene enrichment analysis, pathway enrichment analysis and immune cell deconvolution studies. Consistent with previous studies, our data showed that responders to anti-PD1 therapy had higher immune scores (median immune score for responders = 0.1335, median immune score for non-responders = 0.05426, p-value = 0.01, Mann-Whitney U two-tailed exact test) compared to the non-responders. Responder melanomas were more highly enriched with a combination of CD8+ T cells, dendritic cells (p-value = 0.03) and an M1 subtype of macrophages (p-value = 0.001). In addition, melanomas from responder patients exhibited a more differentiated gene expression pattern, with high proliferative- and low invasive-associated gene expression signatures, whereas tumours from non-responders exhibited high invasive- and frequently neural crest-like cell type gene expression signatures. Our findings suggest that non-responder melanomas to anti-PD1 therapy exhibit a de-differentiated gene expression signature, associated with poorer immune cell infiltration, which establishes a gene expression pattern characteristic of innate resistance to anti-PD1 therapy. Improved understanding of tumour-intrinsic gene expression patterns associated with response to anti-PD1 therapy will help to identify predictive biomarkers of ICI response and may help to identify new targets for anticancer treatment, especially with a capacity to function as adjuvants to improve ICI outcomes.
Collapse
Affiliation(s)
- Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Gregory Gimenez
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter A. Stockwell
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Cristin G. Print
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Janusz Rys
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland
| | - Bozena Cybulska-Stopa
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland
| | - Magda Ratajska
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Harazin-Lechowska
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland
| | - Suzan Almomani
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Christopher Jackson
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- *Correspondence: Michael R. Eccles,
| |
Collapse
|
38
|
Luo L, Wei Q, Xu C, Dong M, Zhao W. Immune landscape and risk prediction based on pyroptosis-related molecular subtypes in triple-negative breast cancer. Front Immunol 2022; 13:933703. [PMID: 36189269 PMCID: PMC9524227 DOI: 10.3389/fimmu.2022.933703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
The survival outcome of triple-negative breast cancer (TNBC) remains poor, with difficulties still existing in prognosis assessment and patient stratification. Pyroptosis, a newly discovered form of programmed cell death, is involved in cancer pathogenesis and progression. The role of pyroptosis in the tumor microenvironment (TME) of TNBC has not been fully elucidated. In this study, we disclosed global alterations in 58 pyroptosis-related genes at somatic mutation and transcriptional levels in TNBC samples collected from The Cancer Genome Atlas and Gene Expression Omnibus databases. Based on the expression patterns of genes related to pyroptosis, we identified two molecular subtypes that harbored different TME characteristics and survival outcomes. Then, based on differentially expressed genes between two subtypes, we established a 12-gene score with robust efficacy in predicting short- and long-term overall survival of TNBC. Patients at low risk exhibited a significantly better prognosis, more antitumor immune cell infiltration, and higher expression of immune checkpoints including PD-1, PD-L1, CTLA-4, and LAG3. The comprehensive analysis of the immune landscape in TNBC indicated that alterations in pyroptosis-related genes were closely related to the formation of the immune microenvironment and the intensity of the anticancer response. The 12-gene score provided new information on the risk stratification and immunotherapy strategy for highly heterogeneous patients with TNBC.
Collapse
|
39
|
Xiang X, Li Y, Yang X, Guo W, Zhou P. Clinical utility of tumour mutational burden on efficacy of immune checkpoint inhibitors in malignant solid tumours: protocol for a systematic review and meta-analysis. BMJ Open 2022; 12:e058692. [PMID: 35926995 PMCID: PMC9358952 DOI: 10.1136/bmjopen-2021-058692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION A major development in solid malignancy treatment is the application of immune checkpoint inhibitors (ICIs), which have produced durable responses and increased survival rates. However, the therapeutic effect of ICIs has great heterogeneity in patients with cancer. We propose a systematic review to evaluate the predictive value of tumour mutation burden (TMB) on efficacy of ICIs. METHODS AND ANALYSIS A systematic literature search will be conducted in the PubMed, OVID, Web of Science, Embase and Cochrane Central Register of Controlled Trials Library databases up to 31 May 2022. We will compare the efficacy of ICIs between TMB high group and TMB low group in terms of the HRs of overall survival (OS) and progression-free survival (PFS), and the OR of the objective response rate/overall response rate (ORR). The HRs of PFS and OS, and the OR of ORR, will be measured by an inverse variance weighted fixed effects model (I2≤50%) or a DerSimonian-Laird random effects model (I2>50%). In addition, subgroup analysis, sensitivity analysis, heterogeneity analysis and publication bias will be conducted. We plan to conduct a subgroup analysis on age, sex, area, number of patients (high/low TMB), cancer type, tumour size, stage, line of therapy, TMB sequencing method, type of immunotherapy and follow-up period. ETHICS AND DISSEMINATION Ethical approval and informed consent are not needed, as the study will be a literature review and will not involve direct contact with patients or alterations to patient care. This systematic review is anticipated to be finished in December 2023, and the results will be published in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42021262480.
Collapse
Affiliation(s)
- Xuemei Xiang
- Basic Medical Laboratory, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yunming Li
- Department of Information, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Department of Statistics, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xiaoguang Yang
- Department of Information, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wang Guo
- Department of Information, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Department of Statistics, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Pengfei Zhou
- Department of Information, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|