1
|
He S, Sun S, Liu K, Pang B, Xiao Y. Comprehensive assessment of computational methods for cancer immunoediting. CELL REPORTS METHODS 2025; 5:101006. [PMID: 40132544 PMCID: PMC12049729 DOI: 10.1016/j.crmeth.2025.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/23/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Cancer immunoediting reflects the role of the immune system in eliminating tumor cells and shaping tumor immunogenicity, which leaves marks in the genome. In this study, we systematically evaluate four methods for quantifying immunoediting. In colorectal cancer samples from The Cancer Genome Atlas, we found that these methods identified 78.41%, 46.17%, 36.61%, and 4.92% of immunoedited samples, respectively, covering 92.90% of all colorectal cancer samples. Comparison of 10 patient-derived xenografts (PDXs) with their original tumors showed that different methods identified reduced immune selection in PDXs ranging from 44.44% to 60.0%. The proportion of such PDX-tumor pairs increases to 77.78% when considering the union of results from multiple methods, indicating the complementarity of these methods. We find that observed-to-expected ratios highly rely on neoantigen selection criteria and reference datasets. In contrast, HLA-binding mutation ratio, immune dN/dS, and enrichment score of cancer cell fraction were less affected by these factors. Our findings suggest integration of multiple methods may benefit future immunoediting analyses.
Collapse
Affiliation(s)
- Shengyuan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Shangqin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
2
|
Stiegeler N, Garsed DW, Au-Yeung G, Bowtell DDL, Heinzelmann-Schwarz V, Zwimpfer TA. Homologous recombination proficient subtypes of high-grade serous ovarian cancer: treatment options for a poor prognosis group. Front Oncol 2024; 14:1387281. [PMID: 38894867 PMCID: PMC11183307 DOI: 10.3389/fonc.2024.1387281] [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: 02/17/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Approximately 50% of tubo-ovarian high-grade serous carcinomas (HGSCs) have functional homologous recombination-mediated (HR) DNA repair, so-called HR-proficient tumors, which are often associated with primary platinum resistance (relapse within six months after completion of first-line therapy), minimal benefit from poly(ADP-ribose) polymerase (PARP) inhibitors, and shorter survival. HR-proficient tumors comprise multiple molecular subtypes including cases with CCNE1 amplification, AKT2 amplification or CDK12 alteration, and are often characterized as "cold" tumors with fewer infiltrating lymphocytes and decreased expression of PD-1/PD-L1. Several new treatment approaches aim to manipulate these negative prognostic features and render HR-proficient tumors more susceptible to treatment. Alterations in multiple different molecules and pathways in the DNA damage response are driving new drug development to target HR-proficient cancer cells, such as inhibitors of the CDK or P13K/AKT pathways, as well as ATR inhibitors. Treatment combinations with chemotherapy or PARP inhibitors and agents targeting DNA replication stress have shown promising preclinical and clinical results. New approaches in immunotherapy are also being explored, including vaccines or antibody drug conjugates. Many approaches are still in the early stages of development and further clinical trials will determine their clinical relevance. There is a need to include HR-proficient tumors in ovarian cancer trials and to analyze them in a more targeted manner to provide further evidence for their specific therapy, as this will be crucial in improving the overall prognosis of HGSC and ovarian cancer in general.
Collapse
Affiliation(s)
| | - Dale W. Garsed
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - George Au-Yeung
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - David D. L. Bowtell
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Tibor A. Zwimpfer
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Gynecological Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
3
|
Lv Q, Zhang Z, Fu H, Li D, Liu Y, Sun Y, Wu M. Predictive Panel for Immunotherapy in Low-Grade Glioma. World Neurosurg 2024; 183:e825-e837. [PMID: 38216032 DOI: 10.1016/j.wneu.2024.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND The main treatment of low-grade glioma (LGG) is still surgical resection followed by radiotherapy and/or chemotherapy, which has certain limitations, including side effects and drug resistance. Immunotherapy is a promising treatment for LGG, but it is generally hindered by the tumor microenvironment with the limited expression of tumor antigens. METHODS We integrated RNA sequencing data sets and clinical information and conducted consistent cluster analysis to explore the most suitable patients for immune checkpoint therapy. Gene set enrichment analysis, UMAP analysis, mutation correlation analysis, TIMER analysis, and TIDE analysis were used to identify the immune characteristics of 3 immune subtypes and the feasibility of 5 antigens as immune checkpoint markers. RESULTS We analyzed the isolation and mutation of homologous recombination repair genes (HRR) of the 3 immune subtypes, and the HRR genes of the 3 subtypes were obviously segregated. Among them, the IS2 subtype has a large number of HRR gene mutations, which increases the immunogenicity of tumors-this is consistent with the results of tumor mutation load analysis of 3 immune subtypes. Then we evaluated the immune cell infiltration of immune subtypes and found that IS2 and IS3 subtypes were rich in immune cells. It is worth noting that there are many Treg cells and NK cells in the IS1 subtype. In addition, when analyzing the immune checkpoint gene expression of the 3 subtypes, we found that they were upregulated most in IS2 subtypes compared with other subtypes. Then when we further confirmed the role of immune-related genes in LGG; through TIDE analysis and TISIDB analysis, we obtained 5 markers that can predict the efficacy of ICB in patients with LGG. In addition, we confirmed that they were associated with poor prognosis through survival analysis. CONCLUSIONS We obtained 3 reliable immune subtypes, and patients with the IS2 subtype are suitable for immunotherapy, in which NAMPT, SLC11A1, TNC, VIM, and SPP1 are predictive panel markers for ICB in the LGG group. Our findings provide a rationale for immunotherapy selection and prediction of patient prognosis in LGG patients.
Collapse
Affiliation(s)
- Qingqing Lv
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyu Zhang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Haijuan Fu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Danyang Li
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yihao Liu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingnan Sun
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Manthopoulou E, Ramai D, Dhar J, Samanta J, Ioannou A, Lusina E, Sacco R, Facciorusso A. Cholangiocarcinoma in the Era of Immunotherapy. Vaccines (Basel) 2023; 11:1062. [PMID: 37376451 PMCID: PMC10301507 DOI: 10.3390/vaccines11061062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/27/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a rare malignancy of the gastrointestinal tract, with aggressive behavior, and portends a poor prognosis. Traditionally, it is classified according to its site of involvement as intrahepatic, perihilar, and distal cholangiocarcinoma. A host of genetic and epigenetic factors have been involved in its pathogenesis. Chemotherapy has remained the standard first-line treatment over the last decade, with a disappointing median overall survival of 11 months for locally advanced and metastatic CCA. The advent of immunotherapy has revolutionized the treatment of many pancreaticobiliary malignancies, offering durable responses with a safe therapeutic profile. To date, there have been no significant advances in the management of CCA. Novel immunotherapeutic methods, such as cancer vaccines, adoptive cell therapy, and combinations of immune checkpoint inhibitors with other agents, are currently under investigation and may improve prognosis with overall survival. Efforts to find robust biomarkers for response to treatment along with multiple clinical trials are also ongoing in this regard. In this review, we present an overview of the current advances and the future perspectives of immunotherapy in the management of CCA.
Collapse
Affiliation(s)
- Eleni Manthopoulou
- Department of Gastroenterology, St. Savvas Oncology Hospital of Athens, 11522 Athens, Greece;
| | - Daryl Ramai
- Gastroenterology and Hepatology, University of Utah Health, Salt Lake City, UT 801385, USA;
| | - Jahnvi Dhar
- Department of Gastroenterology, Sohana Multi-Speciality Hospital, Mohali 140308, India;
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India;
| | - Jayanta Samanta
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India;
| | - Alexandros Ioannou
- Department of Gastroenterology, Alexandra General Hospital, Lourou 4-2, 11528 Athens, Greece;
| | - Ekaterina Lusina
- Therapeutic Unit, Gastroenterology Department, Chaika Clinics, Lesnaya Street 9, 125196 Moscow, Russia;
| | - Rodolfo Sacco
- Department Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy;
| | - Antonio Facciorusso
- Department Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy;
| |
Collapse
|
5
|
Deshmukh R, Prajapati M, Harwansh RK. A review on emerging targeted therapies for the management of metastatic colorectal cancers. Med Oncol 2023; 40:159. [PMID: 37097307 DOI: 10.1007/s12032-023-02020-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/08/2023] [Indexed: 04/26/2023]
Abstract
Colorectal cancers are among the most commonly found cancers over the world. In spite of the recent advancements in diagnosis and prognosis, the management of this metastatic condition remains a challenge. The utility of monoclonal antibodies in the healing of patients with colorectal cancer has opened a new chapter in the quest for newer therapies. The resistance to the standard treatment regimen made it mandatory to search for newer targets. Mutagenic alterations in the gene engaged in cellular differentiation and growth pathway have been the reason for resistance to treatment. The newer therapies target the various proteins and receptors involved in the signal transduction and down streaming pathways leading to cell proliferation. This review presents an insight into the newer targeted therapies for colorectal cancer involving tyrosine kinase blockers, epidermal growth factor receptors, vascular endothelial growth factor, immune checkpoint therapy, and BRAF inhibitors.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
| | - Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
6
|
Current Targeted Therapy for Metastatic Colorectal Cancer. Int J Mol Sci 2023; 24:ijms24021702. [PMID: 36675216 PMCID: PMC9864602 DOI: 10.3390/ijms24021702] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/07/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer deaths worldwide. Surgery or surgery plus radiotherapy and/or chemotherapy for patients with metastatic CRC (mCRC) were accepted as the main therapeutic strategies until the early 2000s, when targeted drugs, like cetuximab and bevacizumab, were developed. The use of targeted drugs in clinical practice has significantly increased patients' overall survival. To date, the emergence of several types of targeted drugs has opened new possibilities and revealed new prospects for mCRC treatment. Therapeutic strategies are continually being updated to select the most suitable targeted drugs based on the results of clinical trials that are currently underway. This review discusses the up-to date molecular evidence of targeted therapy for mCRC and summarizes the Food and Drug Administration-approved targeted drugs including the results of clinical trials. We also explain their mechanisms of action and how these affect the choice of a suitable targeted therapy.
Collapse
|
7
|
Zhang L, Wu Z, Li J, Zhu D, Yang L, Shao Y, Lin Y, Liu Z, Cao Y, Zhang G, Shang S, Zhang Y, Wang K. Impact of Homologous Recombination Deficiency on Outcomes in Patients With Triple-Negative Breast Cancer Treated With Carboplatin-Based Neoadjuvant Chemotherapy: Secondary Analysis of the NeoCART Randomized Clinical Trial. JCO Precis Oncol 2023; 7:e2200337. [PMID: 36652665 DOI: 10.1200/po.22.00337] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Pathologic complete response (pCR) rates of patients with triple-negative breast cancer who were administered docetaxel plus carboplatin were significantly higher than those of patients administered epirubicin/cyclophosphamide followed by docetaxel in the neoadjuvant NeoCART trial. Here, we performed a preplanned secondary analysis of the homologous recombination deficiency (HRD) score as a predictor of the pCR in patients with triple-negative breast cancer from the NeoCART cohort. METHODS Pretherapeutic tumor tissues were assessed retrospectively by DNA extraction and sequencing. BRCA1/2 mutations were evaluated in both somatic and germline forms. HRD scores were calculated from genome-wide allele-specific copy number results and comprised telomeric allelic imbalance, loss of heterozygosity, and large-scale state transitions. High HRD scores were defined as ≥ 38, and HRD was defined as either a high HRD score or a deleterious BRCA1/2 mutation. RESULTS HRD testing was completed for 43 (79.6%) of 54 NeoCART cohort patients. Thirty of 43 (69.8%) tumors had high HRD scores, and eight patients had BRCA-mutated tumors. No significant association between BRCA1/2 mutation status and pCR was observed either in the general population or in the two treatment arms. Docetaxel plus carboplatin group patients who achieved pCR had higher HRD scores than non-pCR patients, and this difference approached significance (61.69 ± 24.26 v 39.44 ± 22.83, P = .061). No significant correlations between HRD scores and pCR (61.29 ± 24.02 v 53.21 ± 24.31, P = .480) or residual cancer burden 0/1 (62.50 ± 22.50 v 51.85 ± 24.74, P = .324) were observed in the epirubicin/cyclophosphamide followed by docetaxel group. CONCLUSION HRD is a potential predictive biomarker for clinical benefit from neoadjuvant carboplatin-based chemotherapy and provides a possibility for screening the optimum chemotherapy backbone to combine with immunotherapy.
Collapse
Affiliation(s)
- Liulu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhiyong Wu
- Diagnosis & Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, Guangdong, China
| | - Jie Li
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Dongqin Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Lingling Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Zhenzhen Liu
- Department of Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yin Cao
- Breast Central, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Gangling Zhang
- Breast Surgery, Baotou Cancer Hospital, Baotou, Inner Mongolia, China
| | - Shiyao Shang
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Sato H, Sasaki K, Hara T, Kobayashi S, Doki Y, Eguchi H, Satoh T, Ishii H. Targeting the regulation of aberrant protein production pathway in gastrointestinal cancer treatment. Front Oncol 2022; 12:1018333. [PMID: 36338771 PMCID: PMC9634730 DOI: 10.3389/fonc.2022.1018333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/15/2022] [Indexed: 02/03/2025] Open
Affiliation(s)
- Hiromichi Sato
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuki Sasaki
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shogo Kobayashi
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichiro Doki
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hidetoshi Eguchi
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Taroh Satoh
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
9
|
Factors associated with treatment response to CD19 CAR-T therapy among a large cohort of B cell acute lymphoblastic leukemia. Cancer Immunol Immunother 2021; 71:689-703. [PMID: 34365516 DOI: 10.1007/s00262-021-03009-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T cell therapy has demonstrated striking responses among B cell acute lymphoblastic leukemia (B-ALL), but analyses of potential factors associated with poor response and relapse are lacking. Here, we summarize the long-term follow-up of 254 B-ALL treated with CD19 CAR-T cells from 5 clinical trials (NCT03173417, NCT02546739, and NCT03671460 retrospectively registered on May 23, 2017, March 1, 2018, and September 7, 2018, respectively, at www.clinicaltrials.gov ; ChiCTR-ONC-17012829, and ChiCTR1800016541 retrospectively registered on September 28, 2017, and June 7, 2018, at www.chictr.org.cn ). Our data showed that TP53 mutation, bone marrow blasts > 20%, prior CAR-T/blinatumomab treatment, and severe cytokine release syndrome (CRS) were associated with a lower complete remission (CR) rate while age, extramedullary disease, complex cytogenetics, history of prior transplant, prior courses of chemotherapy, CAR-T cell dose, and manufacturing source of the cellular product did not affect patients' CR rate. Risk factors related to leukemia-free survival (LFS) and overall survival (OS) were history of prior transplant, complex cytogenetics, TP53 mutation, severe CRS, neurotoxicity, and CAR-T therapy without consolidative allogeneic hematopoietic stem cell transplantation (allo-HSCT). Age and CAR-T cell dose did not influence LFS and OS. Patients with consolidative allo-HSCT after CAR-T therapy had a superior OS and LFS compared to those who did not. This benefit was also observed in both pediatric and adult patients as well as in patients either in high- or low-risk groups. This large study to identify risk factors of CR, LFS, and OS may help to maximize clinical outcomes of CAR-T therapy. Précis TP53 mutation and BM blasts > 20% are two independent factors associated with the CR rate. Patients with high tumor burden as well as those with bone marrow blasts < 5% can benefit from consolidative allo-HSCT post-CAR-T therapy.
Collapse
|
10
|
Bagchi A, Beddows I, Cornelius A, Robinson GW, Jewell SD. Rare cases of medulloblastoma with hypermutation. Cancer Rep (Hoboken) 2021; 5:e1521. [PMID: 34351088 PMCID: PMC9124508 DOI: 10.1002/cnr2.1521] [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: 05/26/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Medulloblastoma is the most common malignant brain tumor of childhood and is considered a tumor with low mutational burden (~1 Mut/Mb). Therefore, though the medulloblastoma genomes have been extensively characterized in literature, reports on potential hypermutations and underlying mutagenic processes in medulloblastomas are limited. AIM In this report, we studied the landscape of mutational burden in primary and recurrent medulloblastoma. Furthermore, we wanted to understand the differences in underlying mutagenic mechanisms in medulloblastoma with low and high mutational burdens. METHODS Fifty-three primary and recurrent medulloblastoma genomic sequence were downloaded from the European Genome Archive as BAM files. Thirty-three cases were obtained from formalin-fixed paraffin-embedded tissues from pathology diagnostic archives of Spectrum Health and Cooperative Human Tissue Network. Somatic mutations were called using Mutect2, following best practices guidelines for Genome Analysis Toolkit V4. Mutational signatures were analyzed using deconstructSigs. RESULTS We identified nine medulloblastoma cases with high mutational burden (>5 Mut/Mb). Of them, five cases met the criteria of hypermutation (>10Mut/Mb), two of the five tumors had canonical mutations in the POLE proof-reading domain, where a large proportion of mutations in these tumor genomes contributed to signature 10. The hypermutated cases also demonstrated mutational signatures 14, 15, and 21, indicating the role of mis match repair deficiency in their mutagenesis. Of the four known molecular subgroups in medulloblastoma-SHH, WNT, Group 3, and Group 4-both the POLE-mutated cases belonged to the SHH subgroup. This report identifies rare cases of hypermutation in medulloblastoma driven by defects in DNA repair mechanisms. CONCLUSION Hypermutation in medulloblastoma can impact therapeutic decisions, especially at recurrence in otherwise fatal high risk SHH-medulloblastomas. A defect in DNA repair leading to SHH -medulloblastoma is yet another important mechanism that should be further investigated in the genesis of these tumors. Therefore, this report provides important scientific and clinical rationale for future research looking for incidence of hypermutation in large cohorts of medulloblastoma patients.
Collapse
Affiliation(s)
- Aditi Bagchi
- Van Andel Institute Graduate School, St. Jude Children's Research Hospital, Spectrum Health Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA.,Division of Pediatric Hematology and Oncology, Spectrum Health Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA.,Division of Neuro-oncology, Department of Oncology, St. Jude Children's Hospital Research Hospital, Memphis, Tennessee, USA
| | - Ian Beddows
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Albert Cornelius
- Division of Pediatric Hematology and Oncology, Spectrum Health Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA
| | - Giles W Robinson
- Division of Neuro-oncology, Department of Oncology, St. Jude Children's Hospital Research Hospital, Memphis, Tennessee, USA
| | - Scott D Jewell
- Van Andel Institute Graduate School, St. Jude Children's Research Hospital, Spectrum Health Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA.,Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
11
|
Badve SS, Penault-Llorca F, Reis-Filho JS, Deurloo R, Siziopikou KP, D'Arrigo C, Viale G. Determining PD-L1 Status in Patients with Triple-Negative Breast Cancer: Lessons Learned from IMpassion130. J Natl Cancer Inst 2021; 114:664-675. [PMID: 34286340 DOI: 10.1093/jnci/djab121] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 07/17/2021] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 12% to 17% of all breast cancers and has an aggressive clinical behavior. Increased tumor-infiltrating lymphocyte counts are prognostic for survival in TNBC, making this disease a potential target for cancer immunotherapy (CIT). Research on immunophenotyping of tumor-infiltrating lymphocytes is revealing molecular and structural organization in the tumor microenvironment that may predict patient prognosis. The anti-programmed death-ligand 1 (PD-L1) antibody atezolizumab plus nab-paclitaxel was the first CIT combination to demonstrate progression-free survival benefit and clinically meaningful overall survival benefit in the first-line treatment of metastatic TNBC (mTNBC) in patients with PD-L1-expressing tumor-infiltrating immune cells (IC) in ≥ 1% of the tumor area. This led to its US and EU approval for mTNBC and US approval of the VENTANA PD-L1 (SP142) assay as a companion diagnostic immunohistochemistry (IHC) assay. Subsequently, the anti- programmed death-1 (PD-1) antibody pembrolizumab plus chemotherapy was approved by the FDA for mTNBC based on progression-free survival benefit in patients with a combined positive score ≥10 by its concurrently approved 22C3 companion diagnostic assay. Treatment guidelines now recommend PD-L1 testing for patients with mTNBC, and the testing landscape will likely become increasingly complex as new anti-PD-L1/PD-1 agents and diagnostics are approved for TNBC. Integrating PD-L1 testing into current diagnostic workflows for mTNBC may provide more treatment options for these patients. Therefore, it is critical for medical oncologists and pathologists to understand the available assays and their relevance to therapeutic options to develop an appropriate workflow for IHC testing.
Collapse
Affiliation(s)
- Sunil S Badve
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Regula Deurloo
- Oncology Biomarker Development, F. Hoffmann-La Roche, Ltd, ., Basel, Switzerland
| | - Kalliopi P Siziopikou
- Breast Pathology Section, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Giuseppe Viale
- University of Milan, Milan, Italy.,European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
12
|
Morand S, Devanaboyina M, Staats H, Stanbery L, Nemunaitis J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int J Mol Sci 2021; 22:6532. [PMID: 34207103 PMCID: PMC8234871 DOI: 10.3390/ijms22126532] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer response to immunotherapy is limited; however, the evaluation of sensitive/resistant target treatment subpopulations based on stratification by tumor biomarkers may improve the predictiveness of response to immunotherapy. These markers include tumor mutation burden, PD-L1, tumor-infiltrating lymphocytes, homologous recombination deficiency, and neoantigen intratumoral heterogeneity. Future directions in the treatment of ovarian cancer include the utilization of these biomarkers to select ideal candidates. This paper reviews the role of immunotherapy in ovarian cancer as well as novel therapeutics and study designs involving tumor biomarkers that increase the likelihood of success with immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Susan Morand
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | - Monika Devanaboyina
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | - Hannah Staats
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | | | | |
Collapse
|
13
|
Mpakali A, Stratikos E. The Role of Antigen Processing and Presentation in Cancer and the Efficacy of Immune Checkpoint Inhibitor Immunotherapy. Cancers (Basel) 2021; 13:E134. [PMID: 33406696 PMCID: PMC7796214 DOI: 10.3390/cancers13010134] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Recent clinical successes of cancer immunotherapy using immune checkpoint inhibitors (ICIs) are rapidly changing the landscape of cancer treatment. Regardless of initial impressive clinical results though, the therapeutic benefit of ICIs appears to be limited to a subset of patients and tumor types. Recent analyses have revealed that the potency of ICI therapies depends on the efficient presentation of tumor-specific antigens by cancer cells and professional antigen presenting cells. Here, we review current knowledge on the role of antigen presentation in cancer. We focus on intracellular antigen processing and presentation by Major Histocompatibility class I (MHCI) molecules and how it can affect cancer immune evasion. Finally, we discuss the pharmacological tractability of manipulating intracellular antigen processing as a complementary approach to enhance tumor immunogenicity and the effectiveness of ICI immunotherapy.
Collapse
Affiliation(s)
- Anastasia Mpakali
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
| | - Efstratios Stratikos
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15784 Athens, Greece
| |
Collapse
|
14
|
Coleman C, Stoller S, Grotzer M, Stucklin AG, Nazarian J, Mueller S. Pediatric hemispheric high-grade glioma: targeting the future. Cancer Metastasis Rev 2020; 39:245-260. [PMID: 31989507 DOI: 10.1007/s10555-020-09850-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pediatric high-grade gliomas (pHGGs) are a group of tumors affecting approximately 0.85 children per 100,000 annually. The general outcome for these tumors is poor with 5-year survival rates of less than 20%. It is now recognized that these tumors represent a heterogeneous group of tumors rather than one entity. Large-scale genomic analyses have led to a greater understanding of the molecular drivers of different subtypes of these tumors and have also aided in the development of subtype-specific therapies. For example, for pHGG with NTRK fusions, promising new targeted therapies are actively being explored. Herein, we review the clinico-pathologic and molecular classification of these tumors, historical treatments, current management strategies, and therapies currently under investigation.
Collapse
Affiliation(s)
- Christina Coleman
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCSF Benioff Children's Hospital, Oakland, 747 52nd Street, Oakland, CA, 94609, USA
| | - Schuyler Stoller
- Department of Neurology, University of California, San Francisco, 625 Nelson Rising Lane, Box 0663, San Francisco, CA, 94158, USA
| | - Michael Grotzer
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Ana Guerreiro Stucklin
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Javad Nazarian
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sabine Mueller
- Department of Neurology, University of California, San Francisco, 625 Nelson Rising Lane, Box 0663, San Francisco, CA, 94158, USA.
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- Division of Hematology/Oncology, Department of Pediatrics, University of California, San Francisco, 550 16th Street, 4th Floor, San Francisco, CA, 94158, USA.
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, M779, San Francisco, CA, USA.
| |
Collapse
|
15
|
Ramzy GM, Koessler T, Ducrey E, McKee T, Ris F, Buchs N, Rubbia-Brandt L, Dietrich PY, Nowak-Sliwinska P. Patient-Derived In Vitro Models for Drug Discovery in Colorectal Carcinoma. Cancers (Basel) 2020; 12:cancers12061423. [PMID: 32486365 PMCID: PMC7352800 DOI: 10.3390/cancers12061423] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Lack of relevant preclinical models that reliably recapitulate the complexity and heterogeneity of human cancer has slowed down the development and approval of new anti-cancer therapies. Even though two-dimensional in vitro culture models remain widely used, they allow only partial cell-to-cell and cell-to-matrix interactions and therefore do not represent the complex nature of the tumor microenvironment. Therefore, better models reflecting intra-tumor heterogeneity need to be incorporated in the drug screening process to more reliably predict the efficacy of drug candidates. Classic methods of modelling colorectal carcinoma (CRC), while useful for many applications, carry numerous limitations. In this review, we address the recent advances in in vitro CRC model systems, ranging from conventional CRC patient-derived models, such as conditional reprogramming-based cell cultures, to more experimental and state-of-the-art models, such as cancer-on-chip platforms or liquid biopsy.
Collapse
Affiliation(s)
- George M. Ramzy
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Thibaud Koessler
- Department of Oncology, Geneva University Hospitals, 1211 Geneva, Switzerland; (T.K.); (P.-Y.D.)
| | - Eloise Ducrey
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Thomas McKee
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), 1211 Geneva, Switzerland; (T.M.); (L.R.-B.)
| | - Frédéric Ris
- Translational Department of Digestive and Transplant Surgery, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.R.); (N.B.)
| | - Nicolas Buchs
- Translational Department of Digestive and Transplant Surgery, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.R.); (N.B.)
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), 1211 Geneva, Switzerland; (T.M.); (L.R.-B.)
| | - Pierre-Yves Dietrich
- Department of Oncology, Geneva University Hospitals, 1211 Geneva, Switzerland; (T.K.); (P.-Y.D.)
| | - Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; (G.M.R.); (E.D.)
- Translational Research Center in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-379-3352
| |
Collapse
|
16
|
Mohme M, Maire CL, Schliffke S, Joosse SA, Alawi M, Matschke J, Schüller U, Dierlamm J, Martens T, Pantel K, Riethdorf S, Lamszus K, Westphal M. Molecular profiling of an osseous metastasis in glioblastoma during checkpoint inhibition: potential mechanisms of immune escape. Acta Neuropathol Commun 2020; 8:28. [PMID: 32151286 PMCID: PMC7063778 DOI: 10.1186/s40478-020-00906-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/29/2020] [Indexed: 12/22/2022] Open
Abstract
Peripheral metastases of glioblastoma (GBM) are very rare despite the ability of GBM cells to pass through the blood-brain barrier and be disseminated through the peripheral blood. Here, we describe a detailed genetic and immunological characterization of a GBM metastasis in the skeleton, which occurred during anti-PD-1 immune checkpoint therapy. We performed whole genome sequencing (WGS) and 850 K methylation profiling of the primary and recurrent intracranial GBM as well as one of the bone metastases. Copy number alterations (CNA) and mutational profiles were compared to known genomic alterations in the TCGA data base. In addition, immunophenotyping of the peripheral blood was performed. The patient who was primarily diagnosed with IDH-wildtype GBM. After the resection of the first recurrence, progressive intracranial re-growth was again detected, and chemotherapy was replaced by PD-1 checkpoint inhibition, which led to a complete intracranial remission. Two months later MR-imaging revealed multiple osseous lesions. Biopsy confirmed the GBM origin of the skeleton metastases. Immunophenotyping reflected the effective activation of a peripheral T-cell response, with, however, increase of regulatory T cells during disease progression. WGS sequencing demonstrated distinct genomic alterations of the GBM metastasis, with gains along chromosomes 3 and 9 and losses along chromosome 4, 10, and 11. Mutational analysis showed mutations in potentially immunologically relevant regions. Additionally, we correlated tumour-infiltrating lymphocyte and microglia presence to the occurrence of circulating tumour cells (CTCs) in a larger cohort and found a decreased infiltration of cytotoxic T cells in patients positive for CTCs. This study exemplifies that the tumour microenvironment may dictate the response to immune checkpoint therapy. In addition, our study highlights the fact that despite an effective control of intracranial GBM, certain tumour clones have the ability to evade the tumour-specific T-cell response and cause progression even outside of the CNS.
Collapse
|
17
|
Zhang X, Li JJ, Lu PH. Advances in the development of chimeric antigen receptor-T-cell therapy in B-cell acute lymphoblastic leukemia. Chin Med J (Engl) 2020; 133:474-482. [PMID: 31977556 PMCID: PMC7046249 DOI: 10.1097/cm9.0000000000000638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
CD19-targeted chimeric antigen receptor T-cell (CAR-T) therapy is effective in refractory/relapsed (R/R) B-cell acute lymphoblastic leukemia (B-ALL). This review focuses on achievements, current obstacles, and future directions in CAR-T research. A high complete remission rate of 68% to 93% could be achieved after anti-CD19 CAR-T treatment for B-ALL. Cytokine release syndrome and CAR-T-related neurotoxicity could be managed. In view of difficulties collecting autologous lymphocytes, universal CAR-T is a direction to explore. Regarding the high relapse rate after anti-CD19 CAR-T therapy, the main solutions have been developing new targets including CD22 CAR-T, or CD19/CD22 dual CAR-T. Additionally, some studies showed that bridging into transplant post-CAR-T could improve leukemia-free survival. Some patients who did not respond to CAR-T therapy were found to have an abnormal conformation of the CD19 exon or trogocytosis. Anti-CD19 CAR-T therapy for R/R B-ALL is effective. From individual to universal CAR-T, from one target to multi-targets, CAR-T-cell has a chance to be off the shelf in the future.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Jing-Jing Li
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| | - Pei-Hua Lu
- Department of Hematology, Lu Daopei Hospital, Langfang, Hebei 065201, China
- Lu Daopei Institute of Hematology, Beijing 100176, China
| |
Collapse
|
18
|
Pellegrino B, Musolino A, Llop-Guevara A, Serra V, De Silva P, Hlavata Z, Sangiolo D, Willard-Gallo K, Solinas C. Homologous Recombination Repair Deficiency and the Immune Response in Breast Cancer: A Literature Review. Transl Oncol 2020; 13:410-422. [PMID: 31901781 PMCID: PMC6948367 DOI: 10.1016/j.tranon.2019.10.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022] Open
Abstract
The success of cancer immunotherapy with immune checkpoint blockade (ICB) has demonstrated the importance of targeting a preexisting immune response in a broad spectrum of tumors. This is particularly novel and relevant for less immunogenic tumors, such as breast cancer (BC), where the efficacy of ICB was more evident in the triple-negative (TNBC) subtype, in earlier stages, and in association with chemotherapy. Tumors harboring homologous recombination DNA repair (HRR) deficiency (HRD) are supposed to have a higher number of mutations, hence a higher tumor mutational burden, which could potentially make them more sensitive to immunotherapy. However, the mechanisms involved in ICB sensitivity and patient selection are still yet to be defined in BC: whether the innate system could play a role and how the adaptive immunity could be linked with HRR pathways are the two key points of debate that we will discuss in this article. The aim of this review was to close the loop between what was found in clinical trial results so far, go back to laboratory theory and preclinical results and point out what needs to be clarified from now on.
Collapse
Affiliation(s)
- B Pellegrino
- Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy.
| | - A Musolino
- Medical Oncology and Breast Unit, University Hospital of Parma, Parma, Italy
| | - A Llop-Guevara
- Experimental Therapeutics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain
| | - V Serra
- Experimental Therapeutics Group, Vall D'Hebron Institute of Oncology, Barcelona, Spain
| | - P De Silva
- Molecular Immunology Unit, Institut Jules Bordet and Universitè Libre de Bruxelles, Bruxelles, Belgium
| | - Z Hlavata
- Medical Oncology Department, CHR Mons-Hainaut, Mons, Belgium
| | - D Sangiolo
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy
| | - K Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet and Universitè Libre de Bruxelles, Bruxelles, Belgium
| | - C Solinas
- Molecular Immunology Unit, Institut Jules Bordet and Universitè Libre de Bruxelles, Bruxelles, Belgium; Regional Hospital of Valle D'Aosta, Aosta, Italy.
| |
Collapse
|
19
|
Whitman J, Kardosh A, Diaz L, Fong L, Hope T, Onodera C, Joseph N, Le D, Fisher G, Bergsland E. Complete Response and Immune-Mediated Adverse Effects With Checkpoint Blockade: Treatment of Mismatch Repair–Deficient Colorectal Neuroendocrine Carcinoma. JCO Precis Oncol 2019; 3:1-7. [DOI: 10.1200/po.19.00098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Julia Whitman
- University of California, San Francisco, San Francisco, CA
| | | | - Luis Diaz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lawrence Fong
- University of California, San Francisco, San Francisco, CA
| | - Thomas Hope
- University of California, San Francisco, San Francisco, CA
| | | | - Nancy Joseph
- University of California, San Francisco, San Francisco, CA
| | - Dung Le
- Johns Hopkins University, Baltimore, MD
| | | | | |
Collapse
|
20
|
Reynolds IS, O'Connell E, Fichtner M, McNamara DA, Kay EW, Prehn JHM, Furney SJ, Burke JP. Mucinous adenocarcinoma of the colon and rectum: A genomic analysis. J Surg Oncol 2019; 120:1427-1435. [PMID: 31729037 DOI: 10.1002/jso.25764] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES Mucinous adenocarcinoma is a distinct subtype of colorectal cancer (CRC) with a worse prognosis when compared with non-mucinous adenocarcinoma. The aim of this study was to compare somatic mutations and copy number alteration (CNA) between mucinous and non-mucinous CRC. METHODS Data from The Cancer Genome Atlas-colon adenocarcinoma and rectum adenocarcinoma projects were utilized. Mucinous and non-mucinous CRC were compared with regard to microsatellite status, overall mutation rate, the most frequently mutated genes, mutations in genes coding for mismatch repair (MMR) proteins and genes coding for mucin glycoproteins. CNA analysis and pathway analysis was undertaken. RESULTS Mucinous CRC was more likely to be microsatellite instability-high (MSI-H) and hypermutated. When corrected for microsatellite status the single-nucleotide variation and insertion-deletion rate was similar between the two cohorts. Mucinous adenocarcinoma was more likely to have mutations in genes coding for MMR proteins and mucin glycoproteins. Pathway analysis revealed further differences between the two histological subtypes in the cell cycle, RTK-RAS, transforming growth factor-β, and TP53 pathways. CONCLUSIONS Mucinous CRC has some distinct genomic aberrations when compared with non-mucinous adenocarcinoma, many of which are driven by the increased frequency of MSI-H tumors. These genomic aberrations may play an important part in the difference seen in response to treatment and prognosis in mucinous adenocarcinoma.
Collapse
Affiliation(s)
- Ian S Reynolds
- Department of Colorectal Surgery, Beaumont Hospital, Dublin, Ireland.,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Emer O'Connell
- Department of Colorectal Surgery, Beaumont Hospital, Dublin, Ireland.,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael Fichtner
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Deborah A McNamara
- Department of Colorectal Surgery, Beaumont Hospital, Dublin, Ireland.,Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elaine W Kay
- Department of Pathology, Beaumont Hospital, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Simon J Furney
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.,Genomic Oncology Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - John P Burke
- Department of Colorectal Surgery, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
21
|
Bauer J, Nelde A, Bilich T, Walz JS. Antigen Targets for the Development of Immunotherapies in Leukemia. Int J Mol Sci 2019; 20:ijms20061397. [PMID: 30897713 PMCID: PMC6471800 DOI: 10.3390/ijms20061397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023] Open
Abstract
Immunotherapeutic approaches, including allogeneic stem cell transplantation and donor lymphocyte infusion, have significantly improved the prognosis of leukemia patients. Further efforts are now focusing on the development of immunotherapies that are able to target leukemic cells more specifically, comprising monoclonal antibodies, chimeric antigen receptor (CAR) T cells, and dendritic cell- or peptide-based vaccination strategies. One main prerequisite for such antigen-specific approaches is the selection of suitable target structures on leukemic cells. In general, the targets for anti-cancer immunotherapies can be divided into two groups: (1) T-cell epitopes relying on the presentation of peptides via human leukocyte antigen (HLA) molecules and (2) surface structures, which are HLA-independently expressed on cancer cells. This review discusses the most promising tumor antigens as well as the underlying discovery and selection strategies for the development of anti-leukemia immunotherapies.
Collapse
Affiliation(s)
- Jens Bauer
- Department of Hematology and Oncology, University Hospital Tübingen, 72076 Tübingen, Germany.
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany.
| | - Annika Nelde
- Department of Hematology and Oncology, University Hospital Tübingen, 72076 Tübingen, Germany.
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany.
| | - Tatjana Bilich
- Department of Hematology and Oncology, University Hospital Tübingen, 72076 Tübingen, Germany.
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany.
| | - Juliane S Walz
- Department of Hematology and Oncology, University Hospital Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
22
|
Solinas C, Marcoux D, Garaud S, Vitória JR, Van den Eynden G, de Wind A, De Silva P, Boisson A, Craciun L, Larsimont D, Piccart-Gebhart M, Detours V, t'Kint de Roodenbeke D, Willard-Gallo K. BRCA gene mutations do not shape the extent and organization of tumor infiltrating lymphocytes in triple negative breast cancer. Cancer Lett 2019; 450:88-97. [PMID: 30797818 DOI: 10.1016/j.canlet.2019.02.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/23/2018] [Accepted: 02/11/2019] [Indexed: 11/27/2022]
Abstract
This study investigated the prevalence of TIL subpopulations, TLS, PD-1 and PD-L1 in tumors from TNBC patients harboring wild-type or mutated BRCA1 or BRCA2 germline genes. This TNBC cohort included 85% TIL-positive (≥10%) tumors with 21% classified as TILhi (≥50%). Interestingly, the BRCAmut group had a significantly higher incidence of TILpos tumors compared to the BRCAwt group (P = 0.037). T cells were dominant in the infiltrate but no statistically significant differences were detected between BRCAwt and BRCAmut for CD3+, CD4+ and CD8+ T cells or CD20+ B cells. TLS were detected in 74% of tumors but again no significant differences between the BRCA groups. PD-1 expression was observed in 33% and PD-L1 in 53% (any cell, cut-off ≥1%) tumors for the entire TNBC cohort. PD-1 expression correlated with PD-L1 and both with TIL and TLS but was not associated with BRCA mutational status. Our analyses reveal that BRCAwt and BRCAmut TNBC are similar except for a significant increase of TILpos tumors in the BRCAmut group. While BRCA gene mutations may not directly drive immune infiltration, the greater number of TILpos tumors could signal greater immunogenicity in this group.
Collapse
Affiliation(s)
- Cinzia Solinas
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Diane Marcoux
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Soizic Garaud
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | | | - Gert Van den Eynden
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium; Department of Pathology, GZA Ziekenhuizen, Sint-Augustinus Campus, Wilrijk, Belgium.
| | | | - Pushpamali De Silva
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Anaïs Boisson
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Ligia Craciun
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium.
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium.
| | - Martine Piccart-Gebhart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Vincent Detours
- IRIBHM, Bioinformatics Laboratory, Université Libre de Bruxelles, Brussels, Belgium.
| | | | - Karen Willard-Gallo
- Molecular Immunology Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|