1
|
Li A, Gonda BL, Codd EM, von Paternos A, Mitchell DR, Herrmann MD, Kalyan P, Flynn SE, Dzu TQ, Gao C, Zhang E, Mendel JJ, Thierauf JC, Sadow PM, Denize T, Yang D, Fintelmann FJ, Fordham JA, Merkin RD, Bhan AK, Huang YC, Raizer J, Faquin WC, Faden DL, Gao X, Park JC, Wirth LJ, Kaluziak ST, Iafrate AJ. Reversible downregulation of HLA class I in adenoid cystic carcinoma. J Immunother Cancer 2025; 13:e011380. [PMID: 40254393 PMCID: PMC12010351 DOI: 10.1136/jitc-2024-011380] [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: 12/22/2024] [Accepted: 04/03/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) is a rare, but lethal cancer with low response rates to systemic therapies, such as cytotoxic chemotherapy and immune-checkpoint inhibitors (ICIs). Despite extensive clinical trials, no effective treatments for patients with recurrent or metastatic ACC are available, and ACC mortality rates remain poor. METHODS We employed automated multiplex immunofluorescence (mIF), single-cell RNA sequencing (scRNA-seq) Gene Expression analysis, RNA in-situ hybridization, and spatial transcriptomics analysis to characterize the immune landscape of ACC tumors, ACC metastasis, and normal tissues from regions where ACCs arise. Based on results from these studies, we treated freshly resected ACCs with interferon-γ or a stimulator of the interferon genes (STING) agonist in vitro. Additionally, we included one patient with ACC in a phase 1 clinical study of a novel STING agonist (dazostinag) plus pembrolizumab. RESULTS The mIF analysis revealed that ACC tumors are immunologically "cold", with few tumor-infiltrating T-lymphocytes and low programmed death-ligand 1 (PD-L1) expression. The most striking finding was a very low beta-2-microglobulin (B2M) expression in nearly all ACCs, with only focal expression found in some ACC metastases. mIF and RNA sequencing analyses of normal salivary gland and breast tissues revealed a p63+, NFIB+, basal duct cell population, with similarly low B2M/human leukocyte antigen (HLA) class I expression. Spatial transcriptomics analysis of the focally B2M-positive ACC metastases uncovered the genetic pathway driving upregulation of B2M, an interferon-γ program mediating the reintroduction of HLA-I/B2M; the significantly upregulated genes included IRF1, GBP1, and TAP1. On short-term treatment of primary ACC tissues in vitro with interferon-γ or a STING agonist, we observed strongly upregulated HLA class I/B2M expression. Moreover, treatment of a patient with recurrent, metastatic breast ACC with a STING agonist and pembrolizumab led to a partial response with a 70% tumor reduction. CONCLUSIONS Low B2M/HLA class I expression may explain why ACCs are immunologically cold and the lack of response to ICIs. Our findings suggest that the normal cell of ACC origin exists in a B2M/HLA-class I low state, and that pharmacologic manipulation with immune activators, such as STING agonists, can restore HLA/B2M in ACCs, as supported by the promising response observed in a patient with metastatic ACC. These findings indicate a potential path to urgently needed immunotherapies.
Collapse
Affiliation(s)
- Annie Li
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bianca L Gonda
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth M Codd
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam von Paternos
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dawn R Mitchell
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Prinjali Kalyan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha E Flynn
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thuc Q Dzu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chengzhuo Gao
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Edwin Zhang
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julia J Mendel
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Julia C Thierauf
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Denize
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Diane Yang
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian J Fintelmann
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Masachusetts, USA
| | - Jo Anne Fordham
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ross D Merkin
- Department of Medicine, Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Atul K Bhan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yu-Chung Huang
- Oncology Development, Takeda Pharmaceuticals, Development Center Americas Inc, Lexington, Massachusetts, USA
| | - Jeffrey Raizer
- Oncology Development, Takeda Pharmaceuticals, Development Center Americas Inc, Lexington, Massachusetts, USA
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel L Faden
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Xin Gao
- Department of Medicine, Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jong Chul Park
- Department of Medicine, Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lori J Wirth
- Department of Medicine, Hematology-Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan T Kaluziak
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Chae YK, Duan R, Chung LI, Oh Y, Alexiev B, Shin S, Kim S, Helenowski I, Matsangou M, Villaflor V, Mahalingam D. Phase II Study of Nivolumab and Ipilimumab for Treatment of Metastatic/Recurrent Adenoid Cystic Carcinoma (ACC) of all Anatomic Sites of Origin and Other Malignant Salivary Gland Tumors. Cancer Med 2025; 14:e70724. [PMID: 40166913 PMCID: PMC11959299 DOI: 10.1002/cam4.70724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVE Dual checkpoint inhibitor therapy with nivolumab and ipilimumab has been FDA approved for a number of cancer sites. However, its role in the treatment of ACC and non-ACC salivary gland carcinomas (non-ACC SGC) is not well established. METHODS AND ANALYSIS We performed Simon's two-stage prospective single-institution Phase II clinical trial of nivolumab with ipilimumab. Two cohorts were analyzed: patients with metastatic/recurrent ACC and patients with non-ACC SGC. The primary endpoint was median progression-free survival (PFS); secondary endpoints were overall response rate (ORR), overall survival (OS), and toxicity. RESULTS Patient enrollment was prematurely terminated due to funding constraints. In total, 19 patients with ACC and 5 patients with non-ACC SGC were enrolled. The patients with ACC had a median OS of 30.0 months (95% CI 15.3-NR months), a median PFS of 8.3 months (95% CI 5.5-30.0 months), and a disease control rate (DCR) of 53% (10/19). The ORR in the ACC group was 5% (CR 0%, n = 0; confirmed PR 5%, n = 1), with one patient having continued stable disease at the time of trial conclusion. The patients with non-ACC SGC had a median OS of 10.4 months (95% CI 6.21-NR months), a median PFS of 6.21 months (95% CI 2.83-NR months), and a DCR of 40% (2/5). The ORR in this cohort was 0%. CONCLUSION In patients with recurrent or metastatic ACC and non-ACC SGC, the combination of nivolumab with ipilimumab resulted in moderate disease control. Further studies are warranted to validate our findings. TRIAL NUMBER NCT03146650.
Collapse
Affiliation(s)
- Young Kwang Chae
- Department of Hematology and OncologyNorthwestern UniversityChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Richard Duan
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | | | - Youjin Oh
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Borislav Alexiev
- Department of Surgical PathologyNorthwestern UniversityChicagoIllinoisUSA
| | | | | | - Irene Helenowski
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | | | | | - Devalingam Mahalingam
- Department of Hematology and OncologyNorthwestern UniversityChicagoIllinoisUSA
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
3
|
Stawarz K, Durzynska M, Gałązka A, Gorzelnik A, Zwolinski J, Paszkowska M, Bieńkowska-Pluta K, Misiak-Galazka M. Current landscape and future directions of therapeutic approaches for adenoid cystic carcinoma of the salivary glands (Review). Oncol Lett 2025; 29:153. [PMID: 39898287 PMCID: PMC11782928 DOI: 10.3892/ol.2025.14899] [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: 09/18/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
Adenoid cystic carcinoma (ACC) of the salivary glands is the second most common type of salivary gland cancer, and is characterized by a poor prognosis and an unclear pathology. The incidence of ACC is rare, as it accounts for 10-15% of all salivary gland tumors and affects mainly patients aged between 50 and 60 years. The annual incidence rate is estimated to be ~4.5 cases per 100,000 individuals. Due to its rarity and the use of contaminated cell lines in previous investigations, the precise etiological factors underlying ACC remain poorly understood. Current treatment modalities, typically involving surgery with or without postoperative radiotherapy, often prove unsatisfactory due to the potential for local recurrence and delayed distant metastases, which may manifest 3-5 years after treatment and constitute the primary failure of existing therapeutic approaches. The indolent growth pattern, along with perineural and perivascular invasion, is potentially responsible for the delayed onset of metastases. No effective systemic therapy has been established so far. Therefore, the management of ACC represents a significant therapeutic challenge. Exploring the molecular characteristics of ACC, including the reasons behind its propensity for perineural invasion and its potential correlation with the immune system, offers promising strategies for managing ACC and could open up novel pathways for future therapeutic interventions. Currently, the use of immunotherapy in ACC treatment has shown limited effectiveness. While the exact mechanism underlying the lack of response to immunotherapy in ACC remains unknown, the low levels of tumor-infiltrating lymphocytes in these tumors may contribute to this resistance. Therefore, identifying novel targets to enhance the immune response against tumor cells is essential. The present review provides an update on clinical studies and explores novel therapeutic targets that could be effective in the therapeutic management of ACC.
Collapse
Affiliation(s)
- Katarzyna Stawarz
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Monika Durzynska
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Adam Gałązka
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Anna Gorzelnik
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jakub Zwolinski
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Monika Paszkowska
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Karolina Bieńkowska-Pluta
- Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Magdalena Misiak-Galazka
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
4
|
Fu Y, Chen J, Zhu X, Ding M, Wang H, Fu S. Roles and therapeutic potential of the SLC family in prostate cancer-literature review. BMC Urol 2025; 25:32. [PMID: 39966814 PMCID: PMC11837367 DOI: 10.1186/s12894-025-01714-w] [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: 10/01/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in men worldwide. Despite advances in treatment, many patients develop resistance to conventional therapies. Solute carrier (SLC) proteins, as transmembrane transporters, have recently emerged as potential therapeutic targets due to their role in tumor metabolism and progression. This review summarizes the key roles of six SLC proteins in PCa, including their involvement in metabolic reprogramming, regulation of signaling pathways, and effects on the tumor microenvironment. Although targeting of SLC family members in prostate cancer remains an underexplored area, the growing body of evidence suggests that it holds potential for future development.
Collapse
Affiliation(s)
- Yuanzhi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
- Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Junhao Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
| | - Xingcheng Zhu
- Department of Clinical Laboratory, The Second People's Hospital of Qujing City Qujing, Yunnan, China
| | - Mingxia Ding
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China.
| | - Shi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Wuhua District, Kunming, 650101, Yunnan, China.
| |
Collapse
|
5
|
Hu Z, Jin X, Wang J, Sui Q, Yi Y, Zeng D, Chen Z, Wang Q, Yin J, Wang L, Lin Z. Analysis of the Clinicopathological Characteristics, Genetic Phenotypes, and Prognostics of Primary Pulmonary and Bronchial Adenoid Cystic Carcinoma. Thorac Cancer 2025; 16:e15526. [PMID: 39828507 PMCID: PMC11742639 DOI: 10.1111/1759-7714.15526] [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: 08/28/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Primary pulmonary and bronchial adenoid cystic carcinoma (PACC) is a rare, low-grade malignant tumor of the lung. However, the relationship between its clinical features, surgical prognosis, and genetic phenotype has not been fully described. METHODS PACC patient information was collected from the SEER, TCGA, and Zhongshan Hospital, Fudan University (FDZSH) databases. Overall survival (OS) was evaluated using the Kaplan-Meier method. Univariate and multivariate analyses through Cox proportional hazards regression identified risk factors that predicted OS. The limma and matfools packages from R were used to compare the differential genes and mutations between PACC and LUAD, respectively. RESULTS Two hundred and ninety-two patients, 14 patients, and 12 patients with PACC were identified from the SEER, TCGA, and FDZSH databases, respectively. The 3-, 5-, and 10-year OS of the PACC patients were 91.7%, 88.6%, and 85.0%, respectively, compared to 95.8%, 93.9%, and 93.3% for patients who underwent surgery. Race, pathological grade, M stage, regional node examination, and regional node positive were independent prognostic factors for the OS of patients who underwent surgery. The gene map of PACC and lung adenocarcinoma (LUAD) shows significant differences. Common mutations found in lung cancer were almost undetectable in PACC patients, whereas mutations in the NOTCH pathway were more common. TMB levels and PD-1/PD-L1 expressions were also lower in PACC patients. CONCLUSION Our study analyzed the main factors that influence the prognosis of PACC patients undergoing surgery and discovered the unique genetic phenotype of PACC.
Collapse
Affiliation(s)
- Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jian Wang
- Department of Thoracic SurgeryShanghai Xuhui Central HospitalShanghaiChina
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Jiacheng Yin
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Lin Wang
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zongwu Lin
- Department of Thoracic Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
6
|
Wang X, Ma T, Liu H, Zhang S, Yang G, Zhao Y, Kong L, Gao R, Chen X. Heterogeneous immune landscapes and macrophage dynamics in primary and lung metastatic adenoid cystic carcinoma of the head and neck. Front Immunol 2024; 15:1483887. [PMID: 39697346 PMCID: PMC11653016 DOI: 10.3389/fimmu.2024.1483887] [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: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Recurrent or metastatic adenoid cystic carcinoma (ACC) of the head and neck is rare and highly aggressive. Due to the ineffectiveness of immune checkpoint therapies, this study aims to investigate the tumor immune microenvironment of primary tumor tissues and lung metastatic tissues and to comprehend the challenges of immunotherapy. Methods We analyzed RNA sequencing data and constructed immune landscapes from 25 primary tumors and 34 lung metastases. The data were then validated by immunohistochemistry and single-cell sequencing analysis. Results Compared to adjacent normal tissues, both primary and lung metastatic ACC showed low immune infiltration. Lung metastases had higher immune infiltration levels and antigen presentation scores but also higher T cell exclusion and dysfunction scores. Single-cell sequencing data and immunohistochemistry revealed abundant immunosuppressive tumor-associated macrophages in lung metastases. Patients with high M2 macrophage infiltration had shorter lung metastasis-free survival. Discussion Primary and lung metastatic ACC exhibit heterogeneous tumor immune microenvironments. Higher immune cell infiltration in lung metastases is countered by the presence of suppressive tumor-associated macrophages, which may limit effective anti-tumor responses.
Collapse
Affiliation(s)
- Xuelian Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- National Human Diseases Animal Model Resource Center; State Key Laboratory of Respiratory Health and Multimorbidity, National Health Commission (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Tingyao Ma
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongfei Liu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shujing Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Guoliang Yang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yue Zhao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lu Kong
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Ran Gao
- National Human Diseases Animal Model Resource Center; State Key Laboratory of Respiratory Health and Multimorbidity, National Health Commission (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Xiaohong Chen
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Zhou J, Zhao G, Wang S, Li N. Systemic therapy in the management of metastatic or locally recurrent adenoid cystic carcinoma of the salivary glands: a systematic review of the last decade. Br J Cancer 2024; 131:1021-1031. [PMID: 39097677 PMCID: PMC11405853 DOI: 10.1038/s41416-024-02795-4] [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: 12/18/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024] Open
Abstract
IMPORTANCE Recurrent/metastatic adenoid cystic carcinoma (R/M AdCC) presents a clinical challenge with limited treatment options, particularly in the face of unsatisfactory efficacy from current therapeutic approaches. This review underscores the unmet clinical needs in managing R/M AdCC, emphasising the imperative for novel therapeutic strategies to address this critical gap. OBJECTIVE The primary objective of this review is to comprehensively analyse and assess trials investigating therapeutic approaches for R/M AdCC. Emphasis is placed on endpoints such as tumour response rates and progression-free survival. The specific interventions, populations, and outcomes examined in these trials will be detailed to provide a focused and informative systematic review. EVIDENCE REVIEW The systematic search spanned databases, including PubMed, EMBASE, and the Cochrane database of systematic reviews. Employing terms like "Carcinoma, Adenoid Cystic" and "trial," the search focused on English full-text articles from April 1, 2010, to August 9, 2023. Inclusion criteria encompassed studies with patients having R/M AdCC, involving drug interventions. Study quality was assessed using the Newcastle-Ottawa Scale for retrospective studies, Cochrane ROBINS-I tool for non-randomised trials, and the ROB-2 tool for randomised controlled trials. FINDINGS A total of 46 trials involving 1244 patients are included in this review, encompassing a variety of therapeutic approaches for R/M AdCC. Targeted therapies, particularly Apatinib at 500 mg, exhibit efficacy with a 47.1% objective response rate (ORR). Conversely, immunotherapeutic agents demonstrate suboptimal performance, with an overall ORR ranging from 0 to 18%. While Apatinib shows promise, the review underscores the imperative for a thorough exploration of drugs targeting unique mechanisms in the immunologically cold nature of R/M AdCC. CONCLUSIONS AND RELEVANCE Substantial progress in systemic therapy for R/M AdCC is evident, driven by early-phase clinical trials, particularly with promising outcomes in VEGF-2 inhibitors. However, challenges persist, notably in immunotherapy due to the cancer's immunologically cold nature. Ongoing research, prioritising early-stage trials, is crucial, emphasising exploration of emerging therapies like cell therapy and antibody-drug conjugates. Transitioning to Phase III trials is essential for more precise therapeutic insights. Collaborative efforts and a focus on personalised precision medicine are vital for overcoming challenges and advancing our understanding of treatment efficacy in this rare cancer.
Collapse
Affiliation(s)
- Jiawei Zhou
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guo Zhao
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhang Wang
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ning Li
- Clinical Cancer Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Ferrand FR, Even C, Chabrillac E, Thariat J, Fakhry N, Vergez S, Bensadoun RJ, Sarradin V, Digue L. Systemic therapies for salivary gland cancer: Adenoid cystic carcinoma. REFCOR recommendations by the formal consensus method. Eur Ann Otorhinolaryngol Head Neck Dis 2024; 141:286-291. [PMID: 38061943 DOI: 10.1016/j.anorl.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
OBJECTIVE To determine the therapeutic indications for systemic medical treatment in the management of adenoid cystic carcinoma (ACC) according to the clinical situation. MATERIALS AND METHODS The French Network of Rare Head and Neck Tumors (REFCOR) formed a steering group, which drafted a narrative review of the literature published on Medline and proposed recommendations. The level of adherence to the recommendations was then assessed by a rating group, according to the formal consensus method. RESULTS ACCs are rare tumors and there is currently insufficient evidence to indicate chemotherapy at the localized stage. At the metastatic stage, progression is often slow. In case of oligometastatic ACC, local treatment should be discussed. The most often indolent nature of polymetastatic ACC can allow management by active surveillance. Molecular screening is recommended, for abnormalities potentially accessible to targeted therapy. CONCLUSION ACCs are rare tumors for which there are currently few effective medical treatments. It is therefore recommended to include patients in clinical trials.
Collapse
Affiliation(s)
- F-R Ferrand
- Département d'Oncologie Médicale, Institut Gustave-Roussy, Villejuif, France; Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France; French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France
| | - C Even
- Département d'Oncologie Médicale, Institut Gustave-Roussy, Villejuif, France
| | - E Chabrillac
- Département de Chirurgie, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - J Thariat
- Département de Radiothérapie, Centre François-Baclesse, Caen, France
| | - N Fakhry
- Département d'ORL et Chirurgie Cervico-Faciale, Hôpital La Conception, AP-HM, Marseille, France
| | - S Vergez
- Département de Chirurgie, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France; Département de Chirurgie ORL et Cervico-Faciale, CHU Toulouse-Larrey, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - R-J Bensadoun
- Centre de Haute Énergie, Clinique Saint-Georges, Nice, France
| | - V Sarradin
- Département d'Oncologie Médicale, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France.
| | - L Digue
- Département d'Oncologie Médicale, Hôpital Saint-André, Bordeaux, France
| |
Collapse
|
9
|
Jia Y, Liu Y, Yang H, Yao F. Adenoid cystic carcinoma: insights from molecular characterization and therapeutic advances. MedComm (Beijing) 2024; 5:e734. [PMID: 39263605 PMCID: PMC11387731 DOI: 10.1002/mco2.734] [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: 03/06/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
Adenoid cystic carcinoma (ACC) is a malignant tumor primarily originating from the salivary glands, capable of affecting multiple organs. Although ACC typically exhibits slow growth, it is notorious for its propensity for neural invasion, local recurrence, and distant metastasis, making it a particularly challenging cancer to treat. The complexity of ACC's histological and molecular features poses significant challenges to current treatment modalities, which often show limited effectiveness. Recent advancements in single-cell RNA-sequencing (scRNA-seq) have begun to unravel unprecedented insights into the heterogeneity and subpopulation diversity within ACC, revealing distinct cellular phenotypes and origins. This review delves into the intricate pathological and molecular characteristics of ACC, focusing on recent therapeutic advancements. We particularly emphasize the insights gained from scRNA-seq studies that shed light on the cellular landscape of ACC, underscoring its heterogeneity and pathobiology. Moreover, by integrating analyses from public databases, this review proposes novel perspectives for advancing treatment strategies in ACC. This review contributes to the academic understanding of ACC by proposing novel therapeutic approaches informed by cutting-edge molecular insights, paving the way for more effective, personalized therapeutic approaches for this challenging malignancy.
Collapse
Affiliation(s)
- Yunxuan Jia
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yupeng Liu
- Department of Thoracic Surgery Tumor Hospital Affiliated to Nantong University Nantong Tumor Hospital Nantong China
| | - Haitang Yang
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Feng Yao
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
10
|
Hoff CO, Manzi J, Lazar Neto F, Ferrarotto R. Vascular Endothelial Growth Factor Receptor Inhibitors for Recurrent or Metastatic Adenoid Cystic Carcinoma: A Systematic Review and Meta-Analysis. JAMA Otolaryngol Head Neck Surg 2024; 150:587-597. [PMID: 38814585 PMCID: PMC11140580 DOI: 10.1001/jamaoto.2024.1177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/02/2024] [Indexed: 05/31/2024]
Abstract
Importance There is no systemic therapy for recurrent or metastatic adenoid cystic carcinoma (ACC) approved by the US Food and Drug Administration. Objective To examine the efficacy, safety, and tolerability of vascular endothelial growth factor receptor (VEGFR) inhibitors in recurrent or metastatic ACC. Data Sources PubMed, Embase, and Cochrane Library were systematically searched for studies of VEGFR inhibitors in recurrent or metastatic ACC from database inception to August 31, 2023. Study Selection Inclusion criteria were prospective clinical trials of recurrent or metastatic ACC treated with VEGFR inhibitors, reporting at least 1 outcome of interest specifically for ACC. Of 1963 identified studies, 17 (0.9%) met inclusion criteria. Data Extraction and Synthesis The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) reporting guideline was followed to extract data. Data were pooled using a random-effects generalized linear mixed model with 95% CIs. Main Outcomes and Measures The primary efficacy outcome was best overall response to VEGFR inhibitors, including objective response, stable disease, or progressive disease (PD). Safety and tolerability outcomes included incidence of grade 3 or higher adverse events, rates of exit from trial due to PD or drug-related toxic effects, and dose reduction rate (DRR). Results A total of 17 studies comprising 560 patients with recurrent or metastatic ACC treated with 10 VEGFR inhibitors were included. The objective response rate was 6% (95% CI, 3%-12%; I2 = 71%) and stable disease was the most frequent best overall response (82%; 95% CI, 74%-87%; I2 = 67%). The 6-month disease control (defined as objective response and stable disease) rate was 54% (95% CI, 45%-62%; I2 = 52%). The rate of grade 3 or higher adverse events was 53% (95% CI, 42%-64%; I2 = 81%) and of DRR was 59% (95% CI, 40%-76%). Most patients (57%; 95% CI, 44%-70%; I2 = 83%) continued therapy until PD; 21% (95% CI, 15%-28%; I2 = 62%) of patients suspended therapy for toxic effects. In subgroup analysis by specific VEGFR inhibitor, the objective response rate was 14% (95% CI, 7%-25%; I2 = 0%), stable disease rate was 76% (95% CI, 63%-85%; I2 = 0%), proportion treated until PD was 61% (95% CI, 14%-94%; I2 = 94%), and DRR was 78% (95% CI, 66%-87%; I2 = 39%) with lenvatinib. Corresponding axitinib results were objective response rate of 8% (95% CI, 4%-15%; I2 = 0%) and stable disease rate of 85% (95% CI, 72%-92%; I2 = 69%), with 73% (95% CI, 63%-82%; I2 = 0%) of patients treated until PD, and the DRR was 22% (95% CI, 12%-38%; I2 = 77%). Rivoceranib had the highest objective response rate (24%; 95% CI, 7%-57%) but high heterogeneity among studies (I2 = 95%) and the lowest rate of patients who continued therapy until PD (35%; 95% CI, 20%-55%; I2 = 90%). Conclusions and Relevance This systematic review and meta-analysis found that VEGFR inhibitors were associated with high rates of disease stabilization in recurrent or metastatic ACC. Of 10 included VEGFR inhibitors, lenvatinib and axitinib were associated with the best combined and consistent efficacy, safety, and tolerability profiles, substantiating their inclusion in treatment guidelines.
Collapse
Affiliation(s)
- Camilla O. Hoff
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Joao Manzi
- School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Felippe Lazar Neto
- Instituto do Cancer do Estado de Sao Paulo, University of Sao Paulo, Sao Paulo, Brazil
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
11
|
Romanò R, De Felice F, Ferri A, Della Monaca M, Maroldi R, Licitra L, Locati LD, Alfieri S. Adenoid Cystic carcinoma of minor salivary glands (AdCCmSG): a multidisciplinary update. Expert Rev Anticancer Ther 2024; 24:567-580. [PMID: 38832770 DOI: 10.1080/14737140.2024.2357806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Adenoid cystic carcinoma of minor salivary glands (AdCCmSG) represents a 'rarity in the rarity,' posing a clinical challenge in lack of standardized, evidence-based recommendations. At present, AdCCmSG management is mostly translated from major salivary gland cancers (MSGCs). Ideally, AdCCmSG diagnostic-therapeutic workup should be discussed and carried out within a multidisciplinary, high-expertise setting, including pathologists, surgeons, radiation oncologists and medical oncologists. AREAS COVERED The present review provides an overview of epidemiology and pathologic classification. Moreover, the most recent, clinically relevant updates in the treatment of AdCCmSG (Pubmed searches, specific guidelines) are critically discussed, aiming to a better understanding of this rare pathologic entity, potentially optimizing the care process, and offering a starting point for reflection on future therapeutic developments. EXPERT OPINION The management of rare cancers is often hindered by limited data and clinical trials, lack of evidence-based guidelines, and hardly represented disease heterogeneity, which cannot be successfully tackled with a 'one-size-fits-all' approach. Our goal is to address these potential pitfalls, providing an easy-to-use, updated, multidisciplinary collection of expert opinions concerning AdCCmSG management as of today's clinical practice. We will also cover the most promising future perspectives, based on the potential therapeutic targets highlighted within AdCCmSG's molecular background.
Collapse
Affiliation(s)
- Rebecca Romanò
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca De Felice
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Ferri
- Maxillo-Facial Surgery Division, Head and Neck Department, University Hospital of Parma, Parma, Italy
| | - Marco Della Monaca
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
- Oncological and Reconstructive Maxillo-Facial Surgery Unit, Policlinico Umberto I, Rome, Italy
| | - Roberto Maroldi
- Division of Radiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- University of Milan, Milan, Italy
| | - Laura Deborah Locati
- Translational Oncology Unit, Istituto di Ricerca e Cura a carattere scientifico (IRCCS) Istituti Clinici Scientifici (ICS) Maugeri, Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
12
|
Utsumi Y, Nakaguro M, Tada Y, Nagao T. High-grade salivary carcinomas: A current insight on diagnostic pathology and the key to clinical decision making. Semin Diagn Pathol 2024; 41:197-206. [PMID: 38658249 DOI: 10.1053/j.semdp.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
High-grade carcinomas of the salivary glands are a group of several tumor entities with highly malignant histologic appearances, and have an aggressive biological behavior accompanied by poor a prognosis. In general, they require more intensive treatment than low- or intermediate-grade carcinomas. High-grade salivary carcinomas are rare and the microscopic features often overlap between different tumor types, making an appropriate diagnosis challenging in daily practice settings. However, with recent rapid advances in molecular pathology and molecular-targeted therapy in this field, there is a growing need to properly classify tumors, rather than just diagnosing the cases as "high-grade carcinomas". This leads to specific treatment strategies. In this article, we review representative high-grade salivary gland carcinomas, including salivary duct carcinoma and its histologic subtypes, high-grade mucoepidermoid carcinoma, solid-type adenoid cystic carcinoma, and high-grade transformation of low- or intermediate-grade carcinomas, and discuss their differential diagnoses and clinical implications. Other rare entities, such as neuroendocrine carcinoma, NUT carcinoma, and metastatic carcinoma, should also be considered before diagnosing high-grade carcinoma, NOS. Of these tumors, salivary duct carcinoma has received the most attention because of its strong association with androgen deprivation and anti-HER2 therapies. Other tumor-type-specific treatments include anti-TRK therapy for high-grade transformation of secretory carcinoma, but further therapeutic options are expected to be developed in the future. It should be emphasized that detailed histological evaluation with adequate sampling, in addition to the effective use of molecular ancillary tests, is of the utmost importance for a suitable diagnosis.
Collapse
Affiliation(s)
- Yoshitaka Utsumi
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Masato Nakaguro
- Departments of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yuichiro Tada
- Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
13
|
Hanna GJ, Grover P, Elliott A, McGrath J, Xiu J, Sukari A, Johnson JM, Wise-Draper T. Molecular Profiling and the Impact of Treatment on Outcomes in Adenoid Cystic Carcinoma Type I and II. Clin Cancer Res 2024; 30:2225-2232. [PMID: 38416410 DOI: 10.1158/1078-0432.ccr-23-3182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
PURPOSE Adenoid cystic carcinoma (ACC) is an uncommon salivary gland cancer with no approved therapies available to treat advanced, incurable disease. Recent molecular profiling efforts have identified two important subtypes: the more aggressive ACC-I is characterized by Notch pathway alterations and MYC amplification whereas ACC-II demonstrates a more indolent phenotype and TP63 overexpression. EXPERIMENTAL DESIGN This retrospective observational cohort study involved de-identified samples from 438 patients with ACC with tumor samples sent for commercially-available molecular profiling (Caris Life Sciences). Next-generation whole-exome and whole-transcriptomic sequencing was performed on primary and metastatic samples. Immunostaining for PD-L1 and RNA deconvolution (quanTIseq) was used to explore the tumor immune microenvironment (TME). Real-world clinical and survival outcome metrics were extracted from insurance claims data. RESULTS MYC expression was 1.61-fold higher (39.8 vs. 24.7; P < 0.0001) among NOTCH1-mutant ACC-I tumors, whereas MYB/L1 fusion rates were similar among ACC-I/II. The median B-cell fraction in the TME was higher among ACC-II (7.1% vs. 5.8%; P < 0.01), although infiltrating T cells subsets were low among either ACC subgroup (both <1%). When pooling systemic treatment categories, ACC-I patients had worse outcomes with available therapies (HR, 3.06; 95% confidence interval, 1.65-5.68; P < 0.01), with no significant difference in overall survival between ACC-I/II based on chemotherapy or VEGFR tyrosine kinase inhibitor exposure in smaller subsets. CONCLUSIONS We confirmed the previously reported associations with MYC and TP63 in the prognostically relevant subgroups of ACC-I and -II, respectively, and report immunologic differences among these subtypes. Survival outcomes are comparatively worse in ACC-I regardless of treatment type.
Collapse
MESH Headings
- Humans
- Carcinoma, Adenoid Cystic/genetics
- Carcinoma, Adenoid Cystic/pathology
- Carcinoma, Adenoid Cystic/mortality
- Carcinoma, Adenoid Cystic/drug therapy
- Carcinoma, Adenoid Cystic/immunology
- Carcinoma, Adenoid Cystic/therapy
- Male
- Female
- Middle Aged
- Aged
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Salivary Gland Neoplasms/genetics
- Salivary Gland Neoplasms/pathology
- Salivary Gland Neoplasms/mortality
- Salivary Gland Neoplasms/immunology
- Salivary Gland Neoplasms/therapy
- Salivary Gland Neoplasms/drug therapy
- Biomarkers, Tumor/genetics
- Retrospective Studies
- Adult
- Treatment Outcome
- Gene Expression Profiling
- Proto-Oncogene Proteins c-myc/genetics
- Prognosis
- B7-H1 Antigen/genetics
- Receptor, Notch1/genetics
- Gene Expression Regulation, Neoplastic
- Aged, 80 and over
- Mutation
Collapse
Affiliation(s)
| | - Punita Grover
- University of Cincinnati Cancer Center (UCCC), Cincinnati, Ohio
| | | | | | | | - Ammar Sukari
- Karmanos Cancer Center, Wayne State University, Detroit, Michigan
| | - Jennifer M Johnson
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | |
Collapse
|
14
|
Costa RF, de Oliveira CA, Gomes ÁNDM, Lourenço SV, Coutinho-Camillo CM. Molecular Aspects of Mucoepidermoid Carcinoma and Adenoid Cystic Carcinoma of the Salivary Gland. Head Neck Pathol 2024; 18:34. [PMID: 38658430 PMCID: PMC11043314 DOI: 10.1007/s12105-024-01629-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/12/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Salivary gland tumors (SGTs) are rare and highly heterogeneous lesions, making diagnosis a challenging activity. In addition, the small number of studies and samples evaluated difficults the determination of prognosis and diagnosis. Despite the solid advances achieved by research, there is still an intense need to investigate biomarkers for diagnosis, prognosis and that explain the evolution and progression of SGTs. METHODS We performed a comprehensive literature review of the molecular alterations focusing on the most frequent malignant SGTs: mucoepidermoid carcinoma and adenoid cystic carcinoma. RESULTS Due to the importance of biomarkers in the tumorigenenic process, this review aimed to address the mechanisms involved and to describe molecular and biomarker pathways to better understand some aspects of the pathophysiology of salivary gland tumorigenesis. CONCLUSIONS Molecular analysis is essential not only to improve the diagnosis and prognosis of the tumors but also to identify novel driver pathways in the precision medicine scenario.
Collapse
Affiliation(s)
- Raisa Ferreira Costa
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Primeiro andar, São Paulo, 01508-010, Brazil
| | - Carolinne Alves de Oliveira
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Primeiro andar, São Paulo, 01508-010, Brazil
| | - Ágatha Nagli de Mello Gomes
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Primeiro andar, São Paulo, 01508-010, Brazil
| | | | | |
Collapse
|
15
|
Xu Z, Peng C, Zhang Y, Chen L, Zhai J. Adenoid cystic carcinoma of the retromolar pad region: A case report. Int J Surg Case Rep 2024; 117:109567. [PMID: 38522303 PMCID: PMC10973809 DOI: 10.1016/j.ijscr.2024.109567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024] Open
Abstract
INTRODUCTION Adenoid cystic carcinoma (ACC) is one of the most common salivary gland malignancies, mostly occurs in the major and minor salivary glands in the oral and maxillofacial region. The development of ACC in the retromolar pad is extremely rare, which limits establishing proper diagnosis and management. PRESENTATION OF CASE A patient described a 2-month history of finding a mass behind the lower left posterior teeth. Based on the physical examination and radiographic findings, we got an initial impression of a benign mucocele, the nature of which was to be investigated further. Pathological examination of the resected tissue resulted in a diagnosis of ACC. Follow-up visits showed no recurrence during the subsequent 54 months. DISCUSSION In cases with an uncertain diagnosis based on medical history, clinical features and imaging examinations, it is important to proceed carefully with the possibility of a tumor in mind. CONCLUSION ACC in the retromolar pad is rare and can be easily misdiagnosed. Clinical, radiographic, and pathological evidence confirm a definitive diagnosis. Long-term follow-up is important for the full analysis of ACC treatment.
Collapse
Affiliation(s)
- Zixian Xu
- Department of Basic Science of Stomatology, School of Stomatology Kunming Medical University, 1168 Chunrong West Road, Chenggong Zone, Kunming 650500, People's Republic of China
| | - Canbang Peng
- Department of Oral and Maxillofacial Surgery, School of Stomatology Kunming Medical University, 1088 Middle Haiyuan Road, High-Tech Zone, Kunming 650106, People's Republic of China
| | - Yuhao Zhang
- Department of Oral Medicine, Stomatological Center of the First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan Zone, Kunming 650034, People's Republic of China
| | - Lizhong Chen
- Department of Oral Medicine, School of Stomatology Kunming Medical University, 1088 Middle Haiyuan Road, High-Tech Zone, Kunming 650106, People's Republic of China
| | - Jiemei Zhai
- Department of Basic Science of Stomatology, School of Stomatology Kunming Medical University, 1088 Middle Haiyuan Road, High-Tech Zone, Kunming 650106, People's Republic of China.
| |
Collapse
|
16
|
Hoff CO, Dal Lago EA, Siqueira JM, de Sousa LG, El-Naggar AK, Ahnert JR, Ferrarotto R. First Use of AXL Targeting in Metastatic, Refractory, Adenoid Cystic Carcinoma: A Case Report. JCO Precis Oncol 2024; 8:e2300633. [PMID: 38579194 PMCID: PMC11018191 DOI: 10.1200/po.23.00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/14/2024] [Accepted: 02/14/2024] [Indexed: 04/07/2024] Open
Abstract
First use of AXL-targeting in adenoid cystic carcinoma (ACC); with positive results, ACC now included in AXL studies.
Collapse
Affiliation(s)
- Camilla O. Hoff
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Juliana Mota Siqueira
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luana G. de Sousa
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adel K. El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
17
|
Horio Y, Kuroda H, Masago K, Matsushita H, Sasaki E, Fujiwara Y. Current diagnosis and treatment of salivary gland-type tumors of the lung. Jpn J Clin Oncol 2024; 54:229-247. [PMID: 38018262 DOI: 10.1093/jjco/hyad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
Salivary gland-type tumors of the lung are thought to originate from the submucosal exocrine glands of the large airways. Due to their rare occurrence, reports of their study are limited to small-scale or case reports. Therefore, daily clinical practices often require a search for previous reports. In the last 20 years, several genetic rearrangements have been identified, such as MYB::NF1B rearrangements in adenoid cystic carcinoma, CRTC1::MAML2 rearrangements in mucoepidermoid carcinoma, EWSR1::ATF1 rearrangements in hyalinizing clear cell carcinoma and rearrangements of the EWSR1 locus or FUS (TLS) locus in myoepithelioma and myoepithelial carcinoma. These molecular alterations have been useful in diagnosing these tumors, although they have not yet been linked to molecularly targeted therapies. The morphologic, immunophenotypic, and molecular characteristics of these tumors are similar to those of their counterparts of extrapulmonary origin, so clinical and radiologic differential diagnosis is required to distinguish between primary and metastatic disease of other primary sites. However, these molecular alterations can be useful in differentiating them from other primary lung cancer histologic types. The management of these tumors requires broad knowledge of the latest diagnostics, surgery, radiotherapy, bronchoscopic interventions, chemotherapy, immunotherapy as well as therapeutic agents in development, including molecularly targeted agents. This review provides a comprehensive overview of the current diagnosis and treatment of pulmonary salivary gland tumors, with a focus on adenoid cystic carcinoma and mucoepidermoid carcinoma, which are the two most common subtypes.
Collapse
Affiliation(s)
- Yoshitsugu Horio
- Department of Outpatient Services, Aichi Cancer Center Hospital, Nagoya, Japan
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
- Department of Thoracic Surgery, Teikyo University Hospital, Mizonokuchi, Kanagawa-prefecture, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yutaka Fujiwara
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
18
|
Rached L, Saleh K, Casiraghi O, Even C. Salivary gland carcinoma: Towards a more personalised approach. Cancer Treat Rev 2024; 124:102697. [PMID: 38401478 DOI: 10.1016/j.ctrv.2024.102697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/26/2024]
Abstract
Salivary Gland carcinomas (SGCs) are rare tumors accounting for less than 1% of all cancers with 21 histologically diverse subtypes. The rarity of the disease presents a challenge for clinicians to conduct large size randomized controlled trials. Surgery and radiotherapy remain the only curative treatment for localized disease, whereas treatments for recurrent and metastatic disease remain more challenging with very disappointing results for chemotherapy. The different histological subtypes harbor various genetic alterations, some pathognomonic with a diagnostic impact for pathologists in confirming a difficult diagnosis and others with therapeutic implications regardless of the histologic subtype. Current international guidelines urge pathologists to identify androgen receptor status, HER-2 expression that could be determined by immunohistochemistry, and TRK status in patients with non-adenoid cystic salivary gland carcinoma that are eligible to initiate a systemic treatment, in order to offer them available targeted therapies or refer them to clinical trials based on their mutational profile. A more advanced molecular profiling by next generation sequencing would offer a larger panel of molecular alterations with possible therapeutic implications such as NOTCH, PI3K, BRAF, MYB, and EGFR. In the following review, we present the most common genetic alterations in SGCs as well as actionable mutations with the latest available data on therapeutic options and upcoming clinical trials.
Collapse
Affiliation(s)
- Layal Rached
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Khalil Saleh
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Odile Casiraghi
- Department of Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Caroline Even
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Villejuif 94800, France.
| |
Collapse
|
19
|
Perri F, Fusco R, Sabbatino F, Fasano M, Ottaiano A, Cascella M, Marciano ML, Pontone M, Salzano G, Maiello ME, Montano M, Calogero E, D'Aniello R, Maiolino P, Ciardiello F, Zotta A, Alfieri S, Ionna F. Translational Insights in the Landscape of Salivary Gland Cancers: Ready for a New Era? Cancers (Basel) 2024; 16:970. [PMID: 38473330 DOI: 10.3390/cancers16050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Salivary gland carcinomas (SGCs) are rare neoplasms, representing less than 10% of all head and neck tumors, but they are extremely heterogeneous from the histological point of view, their clinical behavior, and their genetics. The guidelines regarding their treatment include surgery in most cases, which can also play an important role in oligometastatic disease. Where surgery cannot be used, systemic therapy comes into play. Systemic therapy for many years has been represented by polychemotherapy, but recently, with the affirmation of translational research, it can also count on targeted therapy, at least in some subtypes of SGCs. Interestingly, in some SGC histotypes, predominant mutations have been identified, which in some cases behave as "driver mutations", namely mutations capable of governing the carcinogenesis process. Targeting these driver mutations may be an effective therapeutic strategy. Nonetheless, it is not always possible to have drugs suitable for targeting driver mutations-and targeting driver mutations is not always accompanied by a clinical benefit. In this review, we will analyze the main mutations predominant in the various histotypes of SGCs.
Collapse
Affiliation(s)
- Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Roberta Fusco
- Medical Oncology Devision, IGEA S.p.A., 80013 Naples, Italy
| | - Francesco Sabbatino
- Medical Oncology Department, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy
| | - Morena Fasano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Maria Luisa Marciano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Monica Pontone
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Giovanni Salzano
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Elena Maiello
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Massimo Montano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Ester Calogero
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Roberta D'Aniello
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Piera Maiolino
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Alessia Zotta
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Franco Ionna
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| |
Collapse
|
20
|
Geng S, Chen L, Lin W, Wan F, Le Z, Hu W, Chen H, Liu X, Huang Q, Zhang H, Lu JJ, Kong L. Exploring the Therapeutic Potential of Triptonide in Salivary Adenoid Cystic Carcinoma: A Comprehensive Approach Involving Network Pharmacology and Experimental Validation. Curr Pharm Des 2024; 30:2276-2289. [PMID: 38910414 DOI: 10.2174/0113816128315277240610052453] [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: 03/23/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Salivary Adenoid Cystic Carcinoma (ACC) is characterized by a highly invasive and slow-growing pattern, and its etiology remains unidentified. Triptonide (TN) has demonstrated efficacy as a pharmacotherapeutic agent against ACC. Nonetheless, the specific targets and mechanism of molecular action underlying the effectiveness of TN in treating ACC have not been elucidated. OBJECTIVES By integrating network pharmacology within laboratory experiments, this research delves into the prospective targets and molecular mechanisms associated with the application of TN in treating ACC. METHODS Initially, pertinent targets associated with TN against ACC were acquired from public databases. Subsequently, a combination of network pharmacology and bioinformatics analysis was utilized to screen the top 10 hub targets and key signal pathways of TN-treating ACC. Finally, in vitro experiments involving various molecular assays were conducted to evaluate the biological phenotypes of cells following TN treatment, encompassing assessments of apoptosis levels, plate migration, and other parameters, thereby validating pivotal genes and pathways. RESULTS A total of 23 pertinent targets for TN in relation to ACC were identified, with the top 10 hub genes being MAPK8, PTGS2, RELA, MAPK14, NR3C1, HDAC1, PPARG, NFKBIA, AR, and PGR. There was a significant correlation between the TNF signaling pathway and the treatment of ACC with TN. In vitro experiments demonstrated that TN treatment elevated RELA phosphorylation while concurrently reducing MAPK14 phosphorylation and inducing G2/M arrest. TN exhibited the ability to enhance the apoptosis rate through increased caspase-3 activity, elevated levels of Reactive Oxygen Species (ROS), mitochondrial dysfunction, and inhibition of cell migration. CONCLUSION There is a potential therapeutic role for TN in the treatment of ACC through the activation of the TNF signaling pathway. Among the identified candidates, MAPK8, HDAC1, PTGS2, RELA, NR3C1, PPARG, NFKBIA, AR, and PGR emerge as the most pertinent therapeutic targets for TN in the context of ACC treatment.
Collapse
Affiliation(s)
- Shikai Geng
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Li Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Ziyu Le
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Huaiyuan Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Xingyu Liu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Qingting Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Haojiong Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| |
Collapse
|
21
|
Vos JL, Burman B, Jain S, Fitzgerald CWR, Sherman EJ, Dunn LA, Fetten JV, Michel LS, Kriplani A, Ng KK, Eng J, Tchekmedyian V, Haque S, Katabi N, Kuo F, Han CY, Nadeem Z, Yang W, Makarov V, Srivastava RM, Ostrovnaya I, Prasad M, Zuur CL, Riaz N, Pfister DG, Klebanoff CA, Chan TA, Ho AL, Morris LGT. Nivolumab plus ipilimumab in advanced salivary gland cancer: a phase 2 trial. Nat Med 2023; 29:3077-3089. [PMID: 37620627 PMCID: PMC11293616 DOI: 10.1038/s41591-023-02518-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023]
Abstract
Salivary gland cancers (SGCs) are rare, aggressive cancers without effective treatments when metastasized. We conducted a phase 2 trial evaluating nivolumab (nivo, anti-PD-1) and ipilimumab (ipi, anti-CTLA-4) in 64 patients with metastatic SGC enrolled in two histology-based cohorts (32 patients each): adenoid cystic carcinoma (ACC; cohort 1) and other SGCs (cohort 2). The primary efficacy endpoint (≥4 objective responses) was met in cohort 2 (5/32, 16%) but not in cohort 1 (2/32, 6%). Treatment safety/tolerability and progression-free survival (PFS) were secondary endpoints. Treatment-related adverse events grade ≥3 occurred in 24 of 64 (38%) patients across both cohorts, and median PFS was 4.4 months (95% confidence interval (CI): 2.4, 8.3) and 2.2 months (95% CI: 1.8, 5.3) for cohorts 1 and 2, respectively. We present whole-exome, RNA and T cell receptor (TCR) sequencing data from pre-treatment and on-treatment tumors and immune cell flow cytometry and TCR sequencing from peripheral blood at serial timepoints. Responding tumors universally demonstrated clonal expansion of pre-existing T cells and mutational contraction. Responding ACCs harbored neoantigens, including fusion-derived neoepitopes, that induced T cell responses ex vivo. This study shows that nivo+ipi has limited efficacy in ACC, albeit with infrequent, exceptional responses, and that it could be promising for non-ACC SGCs, particularly salivary duct carcinomas. ClinicalTrials.gov identifier: NCT03172624 .
Collapse
Affiliation(s)
- Joris L Vos
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bharat Burman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Swati Jain
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Conall W R Fitzgerald
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lara A Dunn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James V Fetten
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Loren S Michel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anuja Kriplani
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth K Ng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana Eng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vatche Tchekmedyian
- Department of Medicine, Maine Medical Center-Tufts University School of Medicine, Portland, ME, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine Y Han
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zaineb Nadeem
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Yang
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Raghvendra M Srivastava
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manu Prasad
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte L Zuur
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Otorhinolaryngology Head and Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David G Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Luc G T Morris
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Naito T, Noji R, Kugimoto T, Kuroshima T, Tomioka H, Fujiwara S, Suenaga M, Harada H, Kano Y. The Efficacy of Immunotherapy and Clinical Utility of Comprehensive Genomic Profiling in Adenoid Cystic Carcinoma of Head and Neck. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2111. [PMID: 38138214 PMCID: PMC10745089 DOI: 10.3390/medicina59122111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Adenoid cystic carcinoma (ACC) of the head and neck is generally slow-growing but has a high potential for local recurrence and metastasis to distant organs. There is currently no standard pharmacological treatment for recurrent/metastatic (R/M) ACC, and there are cases in which immune checkpoint inhibitors (ICIs) are administered for ACC according to head and neck squamous cell carcinoma (HNSCC). However, the efficacy of ICIs for ACC remains unclear, and the predictive biomarkers need to be elucidated. Materials and Methods: The Center for Cancer Genomics and Advanced Therapeutics (C-CAT) database enabled the retrospective but nationwide analysis of 263 cases of ACC of the head and neck. Then, we examined and reported four cases of ACC that received ICIs and comprehensive genomic profiling (CGP) in our institution. Results: The C-CAT database revealed that 59 cases out of 263 received ICIs, and the best response was 8% of objective response rate (ORR) and 53% of disease control rate (DCR) (complete response, CR 3%, partial response, PR 5%, stable disease, SD 44%, progressive disease, PD 19%, not evaluated, NE 29%). The tumor mutational burden (TMB) in ACC was lower overall compared to HNSCC and could not be useful in predicting the efficacy of ICIs. Some cases with MYB structural variants showed the response to ICIs in the C-CAT database. A patient with MYB fusion/rearrangement variants in our institution showed long-term stable disease. Conclusions: ICI therapy is a potential treatment option, and the MYB structural variant might be a candidate for predictive biomarkers for immunotherapy in patients with R/M ACC.
Collapse
Affiliation(s)
- Takahiro Naito
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Rika Noji
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takuma Kugimoto
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Takeshi Kuroshima
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Hirofumi Tomioka
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Shun Fujiwara
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Mitsukuni Suenaga
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgical Oncology, Division of Health Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Yoshihito Kano
- Department of Clinical Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
23
|
Desilets A, Ho AL. Nivolumab in salivary gland cancers: confronting the challenges of unlocking the therapeutic benefits of immunotherapy for rare diseases. BMJ ONCOLOGY 2023; 2:e000214. [PMID: 39886514 PMCID: PMC11235025 DOI: 10.1136/bmjonc-2023-000214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Affiliation(s)
- Antoine Desilets
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weil Medical College of Cornell University, New York, New York, USA
| |
Collapse
|
24
|
Locati LD, Ferrarotto R, Licitra L, Benazzo M, Preda L, Farina D, Gatta G, Lombardi D, Nicolai P, Vander Poorten V, Chua MLK, Vischioni B, Sanguineti G, Morbini P, Fonseca I, Sozzi D, Merlotti A, Orlandi E. Current management and future challenges in salivary glands cancer. Front Oncol 2023; 13:1264287. [PMID: 37795454 PMCID: PMC10546333 DOI: 10.3389/fonc.2023.1264287] [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: 07/20/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Salivary gland cancers (SGCs) are rare, accounting for less than 5% of all malignancies of the head and neck region, and are morphologically heterogeneous. The diagnosis is mainly based on histology, with the complementary aid of molecular profiling, which is helpful in recognizing some poorly differentiated, borderline, or atypical lesions. Instrumental imaging defines the diagnosis, representing a remarkable tool in the treatment plan. Ultrasound and magnetic resonance are the most common procedures used to describe the primary tumour. The treatment of SGCs is multimodal and consists of surgery, radiotherapy, and systemic therapy; each treatment plan is, however, featured on the patient and disease's characteristics. On 24 June 2022, in the meeting "Current management and future challenges in salivary gland cancers" many experts in this field discussed the state of the art of SGCs research, the future challenges and developments. After the meeting, the same pool of experts maintained close contact to keep these data further updated in the conference proceedings presented here. This review collects the insights and suggestions that emerged from the discussion during and after the meeting per se.
Collapse
Affiliation(s)
- Laura D. Locati
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | - Renata Ferrarotto
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) National Cancer Institute, Milano, Italy
- University of Milan, Milano, Italy
| | - Marco Benazzo
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Department of Otorhinolaryngology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Preda
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Radiology Institute, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Davide Farina
- Azienda Socio-Sanitaria Territoriale (ASST) Spedali Civili di Brescia, Division of Radiology and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Gemma Gatta
- Evaluative Epidemiology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) National Cancer Institute, Milano, Italy
| | - Davide Lombardi
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Study, Brescia, Italy
| | - Piero Nicolai
- Unit of Otorhinolaryngology - Head and Neck Surgery, University of Study, Padova, Italy
| | - Vincent Vander Poorten
- Otorhinolaryngology-Head and Neck Surgery, Leuven Cancer Institute, University Hospital of Leuven, Leuven, Belgium
- Department of Oncology, Section Head and Neck Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Melvin Lee Kiang Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Barbara Vischioni
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Giuseppe Sanguineti
- Department of Radiotherapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Roma, Italy
| | - Patrizia Morbini
- Unit of Pathology, Department of Molecular Medicine, University of Pavia, Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Isabel Fonseca
- Anatomia Patológica, Instituto Português de Oncologia Francisco Gentil, University of Lisbon, Lisbon, Portugal
| | - Davide Sozzi
- Department of Medicine and Surgery, School of Medicine University of Milano-Bicocca, Monza, Italy
- Maxillofacial Surgery Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Anna Merlotti
- Department of Radiation Oncology, Santa Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy, Pavia, Italy
| |
Collapse
|