1
|
Lin N, Chen S, Zheng Z, Song X. Cost-effectiveness of first-line sintilimab plus chemotherapy versus chemotherapy for advanced esophageal carcinoma in China. Expert Rev Pharmacoecon Outcomes Res 2024:1-9. [PMID: 39327693 DOI: 10.1080/14737167.2024.2410248] [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: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND To evaluate the cost-effectiveness of first-line sintilimab plus chemotherapy versus chemotherapy for advanced esophageal squamous cell carcinoma (ESCC) from the perspective of the Chinese health service system. METHODS A partitioned survival model was constructed to simulate quality-adjusted life years and incremental cost-effectiveness ratios over a patient's lifetime based on a phase III clinical trial. RESULTS Sintilimab plus chemotherapy increased by 0.316 QALY and 0.285 QALY with the additional cost of $5692 and $5269, which led to the ICER of $18000/QALY and $18519/QALY gained in the overall population and the patients with CPS ≥ 10, respectively. CONCLUSIONS Compared with chemotherapy alone, sintilimab may be a cost-effective first-line treatment choice for locally advanced or metastatic ESCC.
Collapse
Affiliation(s)
- Nanlong Lin
- Department of Thoracic surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shiting Chen
- Department of General Surgery, Quangang General Hospital, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhiwei Zheng
- Department of Pharmacy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaobing Song
- Department of Quality Management, Ganzhou Fifth People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
2
|
Hapuarachi B, Danson S, Wadsley J, Muthana M. Exercise to transform tumours from cold to hot and improve immunotherapy responsiveness. Front Immunol 2023; 14:1335256. [PMID: 38149260 PMCID: PMC10749948 DOI: 10.3389/fimmu.2023.1335256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Exercise provides significant health benefits to patients diagnosed with cancer including improved survival outcomes, quality of life and reduced cancer recurrence. Across multiple murine cancer models, aerobic exercise and resistance training has exhibited anti-tumour properties illustrated by inhibited tumour growth, reduced metastatic potential and modulation of the tumour microenvironment to allow the recognition and destruction of cancer cells. Clinical studies have demonstrated the rapid mobilisation and circulatory release of mature lymphoid populations, myokines and cytokines that occurs with exercise along with tumour vasculature normalisation. Tumour microenvironments enriched with immune cells with anti-cancer potential, such as CD8+ T cells, are termed 'hot', whilst those favouring an immunosuppressive environment and lacking in effector immune cells are classed as 'cold'. Pre-clinical evidence suggests exercise training has the potential to reprogramme cold tumours to become hot, although this requires validation in clinical studies. This hot environment could potentiate immunotherapy responsiveness, improving survival outcomes of patients undergoing cancer immunotherapy and allow those with typically cold tumours to benefit from immunotherapy. This review discusses the complex interactions between exercise and cancer, including exercise-induced alterations within the tumour microenvironment and systemic immunity. The potential role exercise may play in improving cancer immunotherapy responsiveness is explored. This review also highlights the need for translational studies exploring the role of exercise in patients with cancer with the potential to widen the spectrum of tumours that derive significant benefit from immunotherapy.
Collapse
Affiliation(s)
- Brindley Hapuarachi
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Sarah Danson
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Jon Wadsley
- Weston Park Cancer Centre, Sheffield Teaching Hospitals National Health Service (NHS) Foundation Trust, Sheffield, United Kingdom
| | - Munitta Muthana
- University Sheffield, Division of Clinical Medicine, Sheffield, United Kingdom
| |
Collapse
|
3
|
Larkin J, Del Vecchio M, Mandalá M, Gogas H, Arance Fernandez AM, Dalle S, Cowey CL, Schenker M, Grob JJ, Chiarion-Sileni V, Marquez-Rodas I, Butler MO, Di Giacomo AM, Middleton MR, Lutzky J, de la Cruz-Merino L, Arenberger P, Atkinson V, Hill AG, Fecher LA, Millward M, Nathan PD, Khushalani NI, Queirolo P, Ritchings C, Lobo M, Askelson M, Tang H, Dolfi S, Ascierto PA, Weber J. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III/IV Melanoma: 5-Year Efficacy and Biomarker Results from CheckMate 238. Clin Cancer Res 2023; 29:3352-3361. [PMID: 37058595 PMCID: PMC10472092 DOI: 10.1158/1078-0432.ccr-22-3145] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 04/16/2023]
Abstract
PURPOSE In the phase III CheckMate 238 study, adjuvant nivolumab significantly improved recurrence-free survival (RFS) and distant metastasis-free survival versus ipilimumab in patients with resected stage IIIB-C or stage IV melanoma, with benefit sustained at 4 years. We report updated 5-year efficacy and biomarker findings. PATIENTS AND METHODS Patients with resected stage IIIB-C/IV melanoma were stratified by stage and baseline programmed death cell ligand 1 (PD-L1) expression and received nivolumab 3 mg/kg every 2 weeks or ipilimumab 10 mg/kg every 3 weeks for four doses and then every 12 weeks, both intravenously for 1 year until disease recurrence, unacceptable toxicity, or withdrawal of consent. The primary endpoint was RFS. RESULTS At a minimum follow-up of 62 months, RFS with nivolumab remained superior to ipilimumab (HR = 0.72; 95% confidence interval, 0.60-0.86; 5-year rates of 50% vs. 39%). Five-year distant metastasis-free survival (DMFS) rates were 58% with nivolumab versus 51% with ipilimumab. Five-year overall survival (OS) rates were 76% with nivolumab and 72% with ipilimumab (75% data maturity: 228 of 302 planned events). Higher levels of tumor mutational burden (TMB), tumor PD-L1, intratumoral CD8+ T cells and IFNγ-associated gene expression signature, and lower levels of peripheral serum C-reactive protein were associated with improved RFS and OS with both nivolumab and ipilimumab, albeit with limited clinically meaningful predictive value. CONCLUSIONS Nivolumab is a proven adjuvant treatment for resected melanoma at high risk of recurrence, with sustained, long-term improvement in RFS and DMFS compared with ipilimumab and high OS rates. Identification of additional biomarkers is needed to better predict treatment outcome. See related commentary by Augustin and Luke, p. 3253.
Collapse
Affiliation(s)
- James Larkin
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | | | - Helen Gogas
- National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | - Ivan Marquez-Rodas
- General University Hospital Gregorio Marañón and CIBERONC, Madrid, Spain
| | | | | | | | - Jose Lutzky
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Luis de la Cruz-Merino
- Hospital Universitario Virgen Macarena, Clinical Oncology Department, University of Seville, Seville, Spain
| | - Petr Arenberger
- Charles University Third Faculty of Medicine and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Victoria Atkinson
- Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
| | | | | | - Michael Millward
- University of Western Australia and Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | | | | | | | | | | | | | - Hao Tang
- Bristol Myers Squibb, Princeton, New Jersey
| | | | - Paolo A. Ascierto
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York
| |
Collapse
|
4
|
Herzog TJ, Hays JL, Barlin JN, Buscema J, Cloven NG, Kong LR, Tyagi NK, Lanneau GS, Long BJ, Marsh RL, Seward SM, Starks DC, Welch S, Moore KN, Konstantinopoulos PA, Gilbert L, Monk BJ, O'Malley DM, Chen X, Dalal R, Coleman RL, Sehouli J. ARTISTRY-7: phase III trial of nemvaleukin alfa plus pembrolizumab vs chemotherapy for platinum-resistant ovarian cancer. Future Oncol 2023; 19:1577-1591. [PMID: 37334673 DOI: 10.2217/fon-2023-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Standard single-agent nonplatinum chemotherapy provides only modest benefit in a small proportion of patients with platinum-resistant/-refractory ovarian cancer, with objective response rates of 6-20% and progression-free survival of ≈3-4 months. Nemvaleukin alfa (nemvaleukin, ALKS 4230) is a novel cytokine designed to capture and expand the therapeutic potential of high-dose interleukin-2 (IL-2) while mitigating its associated toxicity issues. Nemvaleukin preferentially activates cytotoxic CD8+ T cells and natural killer cells with minimal, non-dose-dependent effects on CD4+ regulatory T cells. The global, randomized, open-label, phase III ARTISTRY-7 trial will compare efficacy and safety of nemvaleukin plus pembrolizumab with chemotherapy in patients with platinum-resistant ovarian cancer. The primary end point is investigator-assessed progression-free survival. Clinical Trial Registration: GOG-3063; ENGOT-OV68; NCT05092360 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Thomas J Herzog
- University of Cincinnati Cancer Center, UC College of Medicine, Cincinnati, OH 45267, USA
| | - John L Hays
- Wexner Medical Center & James Cancer Hospital, Ohio State University, Columbus, OH 43210, USA
| | | | | | - Noelle G Cloven
- Texas Oncology - Fort Worth Cancer Center, Fort Worth, TX 76104, USA
| | - Lynn R Kong
- Ventura County Hematology Oncology Specialists, Oxnard, CA 93930, USA
| | | | | | - Beverly J Long
- Sarasota Memorial Healthcare System, Sarasota, FL 34239, USA
| | | | | | | | - Stephen Welch
- London Health Sciences Centre, London, ON, N6A 5A5, Canada
| | - Kathleen N Moore
- College of Medicine, University of Oklahoma, Oklahoma City, OK 73117, USA
| | | | - Lucy Gilbert
- McGill University Health Centre, Women's Health Research Unit, Montréal, QC, H3H 2R9, Canada
| | - Bradley J Monk
- HonorHealth Research Institute, University of Arizona College of Medicine, Creighton University School of Medicine, Phoenix, AZ 85012, USA
| | - David M O'Malley
- Wexner Medical Center & James Cancer Hospital, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | - Jalid Sehouli
- Charité Universitaetsmedizin Berlin Charité Campus Virchow-Klinikum, Berlin, 11017, Germany
| |
Collapse
|
5
|
Boulhen C, AIT SSI S, Benthami H, Razzouki I, Lakhdar A, Karkouri M, Badou A. TMIGD2 as a potential therapeutic target in glioma patients. Front Immunol 2023; 14:1173518. [PMID: 37261362 PMCID: PMC10227580 DOI: 10.3389/fimmu.2023.1173518] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/21/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Among all types of central nervous system cancers, glioma remains the most frequent primary brain tumor in adults. Despite significant advances in immunomodulatory therapies, notably immune checkpoint inhibitors, their effectiveness remains constrained due to glioma resistance. The discovery of TMIGD2 (Transmembrane and Immunoglobulin Domain Containing 2) as an immuno-stimulatory receptor, constitutively expressed on naive T cells and most natural killer (NK) cells, has emerged as an attractive immunotherapy target in a variety of cancers. The expression profile of TMIGD2 and its significance in the overall survival of glioma patients remains unknown. Methods In the present study, we first assessed TMIGD2 mRNA expression using the Cancer Genome Atlas (TCGA) glioma transcriptome dataset (667 patients), followed by validation with the Chinese Glioma Genome Atlas (CGGA) cohort (693 patients). Secondly, we examined TMIGD2 protein staining in a series of 25 paraffin-embedded blocks from Moroccan glioma patients. The statistical analysis was performed using GraphPad Prism 8 software. Results TMIGD2 expression was found to be significantly higher in astrocytoma, IDH-1 mutations, low-grade, and young glioma patients. TMIGD2 was expressed on immune cells and, surprisingly, on tumor cells of glioma patients. Interestingly, our study demonstrated that TMIGD2 expression was negatively correlated with angiogenesis, hypoxia, G2/M checkpoint, and epithelial to mesenchymal transition signaling pathways. We also demonstrated that dendritic cells, monocytes, NK cells, gd T cells, and naive CD8 T cell infiltration correlates positively with TMIGD2 expression. On the other hand, Mantel-Cox analysis demonstrated that increased expression of TMIGD2 in human gliomas is associated with good overall survival. Cox multivariable analysis revealed that TMIGD2 is an independent predictor of a good prognosis in glioma patients. Discussion Taken together, our results highlight the tight implication of TMIGD2 in glioma progression and show its promising therapeutic potential as a stimulatory target for immunotherapy.
Collapse
Affiliation(s)
- Chaimae Boulhen
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Saadia AIT SSI
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Hamza Benthami
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Ibtissam Razzouki
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Department of Neurosurgery, Faculty of Medicine and Pharmacy, University of Hassan II, Casablanca, Morocco
| | - Mehdi Karkouri
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco and Mohammed VI University of Sciences and Health, Casablanca, Morocco
| |
Collapse
|
6
|
Fernandes PD, Magalhães FD, Pereira RF, Pinto AM. Metal-Organic Frameworks Applications in Synergistic Cancer Photo-Immunotherapy. Polymers (Basel) 2023; 15:polym15061490. [PMID: 36987269 PMCID: PMC10053741 DOI: 10.3390/polym15061490] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Conventional cancer therapies, such as radiotherapy and chemotherapy, can have long-term side effects. Phototherapy has significant potential as a non-invasive alternative treatment with excellent selectivity. Nevertheless, its applicability is restricted by the availability of effective photosensitizers and photothermal agents, and its low efficacy when it comes to avoiding metastasis and tumor recurrence. Immunotherapy can promote systemic antitumoral immune responses, acting against metastasis and recurrence; however, it lacks the selectivity displayed by phototherapy, sometimes leading to adverse immune events. The use of metal-organic frameworks (MOFs) in the biomedical field has grown significantly in recent years. Due to their distinct properties, including their porous structure, large surface area, and inherent photo-responsive properties, MOFs can be particularly useful in the fields of cancer phototherapy and immunotherapy. MOF nanoplatforms have successfully demonstrated their ability to address several drawbacks associated with cancer phototherapy and immunotherapy, enabling an effective and low-side-effect combinatorial synergistical treatment for cancer. In the coming years, new advancements in MOFs, particularly regarding the development of highly stable multi-function MOF nanocomposites, may revolutionize the field of oncology.
Collapse
Affiliation(s)
- Pedro D. Fernandes
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Fernão D. Magalhães
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Artur M. Pinto
- LEPABE, Faculdade de Engenharia, Universidade do Porto, Rua Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
7
|
Dias e Silva D, Borba GB, Beal JR, Botrus G, Osawa A, Araújo SEA, Moura F, Guendelmann RAK, Uson Junior PLS. Response to Abemaciclib and Immunotherapy Rechallenge with Nivolumab and Ipilimumab in a Heavily Pretreated TMB-H Metastatic Squamous Cell Lung Cancer with CDKN2A Mutation, PIK3CA Amplification and TPS 80%: A Case Report. Int J Mol Sci 2023; 24:ijms24044209. [PMID: 36835617 PMCID: PMC9963198 DOI: 10.3390/ijms24044209] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Inactivation of the cyclin-dependent kinase inhibitor 2A (CDKN2A) gene is considerably more frequent in squamous cell lung cancer (SqCLC) than in other subtypes of lung cancer and may be a promising target for this histology. Here, we present the course of diagnosis and treatment of a patient with advanced SqCLC, harboring not only CDKN2A mutation but also PIK3CA amplification, Tumor Mutational Burden-High (>10 mutations/megabase), and a Tumor Proportion Score of 80%. After disease progression on multiple lines of chemotherapy and immunotherapy, he responded favorably to treatment with the CDK4/6i Abemaciclib and later achieved a durable partial response to immunotherapy rechallenge with a combination of anti-PD-1 and anti-CTLA-4, nivolumab, and ipilimumab.
Collapse
Affiliation(s)
- Douglas Dias e Silva
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Guilherme Bes Borba
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Juliana Rodrigues Beal
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Gehan Botrus
- Department of Oncology and Hematology, Emory University, Atlanta, GA 30322, USA
| | - Akemi Osawa
- Department of Nuclear Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Sérgio Eduardo Alonso Araújo
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | - Fernando Moura
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| | | | - Pedro Luiz Serrano Uson Junior
- Department of Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Correspondence:
| |
Collapse
|
8
|
Shen Y, Nussbaum YI, Manjunath Y, Hummel JJ, Ciorba MA, Warren WC, Kaifi JT, Papageorgiou C, Cortese R, Shyu CR, Mitchem JB. TBX21 Methylation as a Potential Regulator of Immune Suppression in CMS1 Subtype Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14194594. [PMID: 36230517 PMCID: PMC9558549 DOI: 10.3390/cancers14194594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Cytotoxic T lymphocyte (CTL) infiltration is associated with survival, recurrence, and therapeutic response in colorectal cancer (CRC). Immune checkpoint inhibitor (ICI) therapy, which requires CTLs for response, does not work for most CRC patients. Therefore, it is critical to improve our understanding of immune resistance in this disease. We utilized 2391 CRC patients and 7 omics datasets, integrating clinical and genomic data to determine how DNA methylation may impact survival and CTL function in CRC. Using comprehensive molecular subtype (CMS) 1 patients as reference, we found TBX21 to be the only gene with altered expression and methylation that was associated with CTL infiltration. We found that CMS1 patients with high TBX21 expression and low methylation had a significant survival advantage. To confirm the role of Tbx21 in CTL function, we utilized scRNAseq data, demonstrating the association of TBX21 with markers of enhanced CTL function. Further analysis using pathway enrichment found that the genes TBX21, MX1, and SP140 had altered expression and methylation, suggesting that the TP53/P53 pathway may modify TBX21 methylation to upregulate TBX21 expression. Together, this suggests that targeting epigenetic modification more specifically for therapy and patient stratification may provide improved outcomes in CRC.
Collapse
Affiliation(s)
- Yuanyuan Shen
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Yulia I. Nussbaum
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Yariswamy Manjunath
- Harry S. Truman Memorial Veterans’ Hospital, University of Missouri, Columbia, MO 65211, USA
| | - Justin J. Hummel
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Matthew A. Ciorba
- School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Wesley C. Warren
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
- Department of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Jussuf T. Kaifi
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, University of Missouri, Columbia, MO 65211, USA
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
- School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Christos Papageorgiou
- School of Medicine, University of Missouri, Columbia, MO 65211, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65211, USA
| | - Rene Cortese
- School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Chi-Ren Shyu
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
- College of Engineering, University of Missouri, Columbia, MO 65211, USA
| | - Jonathan B. Mitchem
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veterans’ Hospital, University of Missouri, Columbia, MO 65211, USA
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
- School of Medicine, University of Missouri, Columbia, MO 65211, USA
- Correspondence:
| |
Collapse
|
9
|
Multi-Level Analysis and Identification of Tumor Mutational Burden Genes across Cancer Types. Genes (Basel) 2022; 13:genes13020365. [PMID: 35205408 PMCID: PMC8872466 DOI: 10.3390/genes13020365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
Tumor mutational burden (TMB) is considered a potential biomarker for predicting the response and effect of immune checkpoint inhibitors (ICIs). However, there are still inconsistent standards of gene panels using next-generation sequencing and poor correlation between the TMB genes, immune cell infiltrating, and prognosis. We applied text-mining technology to construct specific TMB-associated gene panels cross various cancer types. As a case exploration, Pearson’s correlation between TMB genes and immune cell infiltrating was further analyzed in colorectal cancer. We then performed LASSO Cox regression to construct a prognosis predictive model and calculated the risk score of each sample for receiver operating characteristic (ROC) analysis. The results showed that the assessment of TMB gene panels performed well with fewer than 500 genes, highly mutated genes, and the inclusion of synonymous mutations and immune regulatory and drug-target genes. Moreover, the analysis of TMB differentially expressed genes (DEGs) suggested that JAKMIP1 was strongly correlated with the gene expression level of CD8+ T cell markers in colorectal cancer. Additionally, the prognosis predictive model based on 19 TMB DEGs reached AUCs of 0.836, 0.818, and 0.787 in 1-, 3-, and 5-year OS models, respectively (C-index: 0.810). In summary, the gene panel performed well and TMB DEGs showed great potential value in immune cell infiltration and in predicting survival.
Collapse
|