1
|
Ruiz-Torres DA, Bryan ME, Hirayama S, Merkin RD, Luciani E, Roberts TJ, Patel M, Park JC, Wirth LJ, Sadow PM, Sade-Feldman M, Stott SL, Faden DL. Spatial characterization of tertiary lymphoid structures as predictive biomarkers for immune checkpoint blockade in head and neck squamous cell carcinoma. Oncoimmunology 2025; 14:2466308. [PMID: 39963988 PMCID: PMC11845054 DOI: 10.1080/2162402x.2025.2466308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
Immune checkpoint blockade (ICB) is the standard of care for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), yet efficacy remains low. The combined positive score (CPS) for PD-L1 is the only biomarker approved to predict response to ICB and has limited performance. Tertiary Lymphoid Structures (TLS) have shown promising potential for predicting response to ICB. However, their exact composition, size, and spatial biology in HNSCC remain understudied. To elucidate the impact of TLS spatial biology in response to ICB, we utilized pre-ICB tumor tissue sections from 9 responders (complete response, partial response, or stable disease) and 11 non-responders (progressive disease) classified via RECISTv1.1. A custom multi-immunofluorescence (mIF) staining assay was applied to characterize tumor cells (pan-cytokeratin), T cells (CD4, CD8), B cells (CD19, CD20), myeloid cells (CD16, CD56, CD163), dendritic cells (LAMP3), fibroblasts (α Smooth Muscle Actin), proliferative status (Ki67) and immunoregulatory molecules (PD1). A machine learning model was employed to measure the effect of spatial metrics on achieving a response to ICB. A higher density of B cells (CD20+) was found in responders compared to non-responders to ICB (p = 0.022). The presence of TLS within 100 µm of the tumor was associated with improved overall (p = 0.04) and progression-free survival (p = 0.03). A multivariate machine learning model identified TLS density as a leading predictor of response to ICB with 80% accuracy. Immune cell densities and TLS spatial location play a critical role in the response to ICB in HNSCC and may potentially outperform CPS as a predictor of response.
Collapse
Affiliation(s)
- Daniel A. Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael E. Bryan
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shun Hirayama
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
| | - Ross D. Merkin
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Evelyn Luciani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Thomas J. Roberts
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Manisha Patel
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Jong C. Park
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Lori J. Wirth
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Peter M. Sadow
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shannon L. Stott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Daniel L. Faden
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
2
|
Wu Z, Lepcha TT, Zhou D, He Z, Fiches GN, Park Y, He J, Chen J, Shanaka K, Oghumu S, Zhao W, Ma A, Ma Q, Zhu J, Santoso NG. Analysis of Head and Neck Cancer scRNA-seq Data Identified PRDM6 Promotes Tumor Progression by Modulating Immune Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641548. [PMID: 40093183 PMCID: PMC11908237 DOI: 10.1101/2025.03.04.641548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a biologically aggressive and heterogeneous group of cancers with limited treatment options for patients who do not respond to standard therapies. While HPV-related HNSCCs tend to show better therapeutic outcomes, we still had limited understanding of the immune mechanisms underlying these cancers. Immune-responsive genes (IRGs) have emerged as critical factors in regulating both tumor progression and immune response. Recent advances in single-cell RNA sequencing (scRNA-seq) and the development of cell-type specific regulon inference tools, such as IRIS3, have provided new insights into the tumor immune microenvironment. In this study, we leveraged the IRIS3 platform to analyze scRNA-seq data from HNSCC patient samples, identifying novel transcription factor (TF)-IRG networks that contribute to tumor proliferation and immune escape. Specifically, we identified PRDM6, a histone methyltransferase, possesses the previously unknown role in promoting tumor cell proliferation by inducing IRG expression. We further demonstrated that HPV viral oncoproteins (E6/E7) oncoproteins up-regulate the PRDM6 expression, which associates PRDM6 with HPV-positive HNSCC.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thurbu Tshering Lepcha
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Dawei Zhou
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Zhixian He
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Guillaume N. Fiches
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Youngmin Park
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jinshan He
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jianwen Chen
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - K.A.S.N Shanaka
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Steve Oghumu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Weiqiang Zhao
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jian Zhu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Netty G. Santoso
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Struckmeier AK, Gosau M, Smeets R. Immunotherapeutic strategies beyond the PD-1/PD-L1 pathway in head and neck squamous cell carcinoma - A scoping review on current developments in agents targeting TIM-3, TIGIT, LAG-3, and VISTA. Oral Oncol 2025; 161:107145. [PMID: 39705929 DOI: 10.1016/j.oraloncology.2024.107145] [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: 10/10/2024] [Revised: 11/24/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) poses a considerable challenge due to its high incidence and mortality rates. Immunotherapy targeting PD-(L)1 emerges as a promising approach for HNSCC, as it has the potential to trigger a broad and long-lasting anti-tumor response. Nevertheless, the effectiveness of immunotherapy encounters hurdles, and only a small proportion of patients benefit, with many eventually experiencing relapse. Consequently, there is a pursuit of strategies to enhance overall treatment outcomes. Understanding the mechanisms driving resistance to PD-(L)1 inhibition and devising strategies to overcome these challenges are vital for advancing more effective treatments. Furthermore, gaining insights into the mechanisms of action and safety profiles of novel combination therapies is critical for their successful adoption in clinical practice. As a result, current research is dedicated to investigating various immunotherapeutic agents beyond the PD-1/PD-L1 axis. This review offers a comprehensive overview of the existing immunotherapy strategies in HNSCC with a focus on TIM-3, TIGIT, LAG-3, and VISTA. The aim is to lay a strong foundation for the continual advancement of therapies for HNSCC.
Collapse
Affiliation(s)
- Ann-Kristin Struckmeier
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Meenu M, Dhiman P, Kumar M, Pradhan P, Pandey S. Real-World Evidence on the Effectiveness of Pembrolizumab in Patients With Recurrent/Metastatic/Unresectable Head and Neck Squamous Cell Cancer: A Systematic Review and Meta-Analysis. Cureus 2025; 17:e76709. [PMID: 39748883 PMCID: PMC11694497 DOI: 10.7759/cureus.76709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2024] [Indexed: 01/04/2025] Open
Abstract
The aim of the review was to systematically review real-world data on the effectiveness and safety of pembrolizumab in recurrent/metastatic/unresectable head and neck squamous cell cancer (HNSCC) patients. Two independent reviewers retrieved the studies separately and simultaneously. PubMed, Embase, Scopus, Web of Science, and Cochrane Central were searched for prospective and retrospective studies on recurrent/metastatic/unresectable HNSCC patients treated with either pembrolizumab monotherapy or pembrolizumab combination therapy published till November 2024. Studies published in the English language and available as full-text articles were included while studies with patients treated with additional chemotherapeutic agents that do not include chemotherapy with fluorouracil, platinum, cetuximab, or radiotherapy were excluded. Outcomes were overall response rate, progression-free survival, overall survival, and adverse events. Forest plots were generated using RevMan software version 5.4 (2020; The Cochrane Collaboration, London, United Kingdom). The ROBINS-I (Risk Of Bias In Non-randomised Studies - of Interventions) tool was used for the assessment of the potential risk of bias and the quality of evidence was synthesized using GRADEpro (Evidence Prime Inc., Hamilton, Canada). We identified 5884 records, removed 2784 duplicate records, and screened 3100 studies. A total of 2931 records were excluded based on title/abstract. Of the remaining 169 articles, nine studies satisfied eligibility criteria and were included for final review. Pooled data suggested that 1006 patients were administered with pembrolizumab monotherapy while 448 patients received pembrolizumab combination therapy. Monotherapy improved the overall response rate compared to combination therapy. No significant difference in progression-free survival and overall survival was found between the two groups. Adverse events were reported less in pembrolizumab monotherapy compared to pembrolizumab combination therapy. The studies were assessed at high risk of bias and graded at low to very low quality of evidence. The study showed some beneficial effects of pembrolizumab monotherapy in recurrent/metastatic/unresectable HNSCC patients in real-world scenarios. However, more studies are required to generate robust evidence.
Collapse
Affiliation(s)
- Meenakshi Meenu
- Pharmacology, All India Institute of Medical Sciences, Bilaspur, Bilaspur, IND
| | - Pravesh Dhiman
- Medical Oncology, All India Institute of Medical Sciences, Bilaspur, Bilaspur, IND
| | - Muninder Kumar
- Radiation Oncology, All India Institute of Medical Sciences, Bilaspur, Bilaspur, IND
| | - Pranita Pradhan
- Indian Council of Medical Research (ICMR) Advanced Center for Evidence Based Child Health, Postgraduate Institute of Medical Education and Research, Chandigarh, IND
| | - Shivam Pandey
- Biostatistics, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| |
Collapse
|
5
|
Cheng G, Chen Y, Xu N, Mao C, Zhang N, Chen M, Yan H, Yang Z, Bie J, Ding L, Wang Z, Yan J, Cariati P, Qin S. Safety and efficacy of mecapegfilgrastim in preventing neutropenia in patients with head and neck cancer: a multicenter, prospective, observational, real-world study. Transl Cancer Res 2024; 13:6895-6904. [PMID: 39816534 PMCID: PMC11730191 DOI: 10.21037/tcr-24-2035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/08/2024] [Indexed: 01/18/2025]
Abstract
Background Mecapegfilgrastim, a long-acting granulocyte colony-stimulating factor, is approved in China for neutropenia prevention. However, data on its safety and efficacy in patients with head and neck cancer remain limited. This study aimed to evaluate the safety and efficacy of mecapegfilgrastim in preventing neutropenia among these patients undergoing chemotherapy, particularly those receiving chemoradiotherapy or chemoimmunotherapy. Methods This prospective, non-interventional, real-world study enrolled patients aged ≥18 years with pathologically or cytohistologically confirmed head and neck cancer who were deemed tolerable to mecapegfilgrastim. The administration of mecapegfilgrastim was determined by the local physicians. Patients were observed for up to four chemotherapy cycles, with follow-up 30±2 days post-final administration. The primary outcome was the incidence of adverse events following mecapegfilgrastim administration. Secondary outcomes included the incidence of grade ≥3 and grade 4 neutropenia [defined as absolute neutrophil count (ANC) <1.0×109/L and ANC <0.5×109/L, respectively], febrile neutropenia (FN), and infections across all chemotherapy cycles. Results Between May 2019 and November 2021, 197 patients were enrolled from 24 sites across China. The median age was 53 years, and 76.6% were male. Among them, 25.9% underwent chemoradiotherapy and 38.6% received chemoimmunotherapy. Treatment-related adverse events (TRAEs) of any grade occurred in 18 (9.1%) of all patients, 6 (11.8%) of the 51 patients who underwent chemoradiotherapy, and 4 (5.3%) of the 76 patients who received chemoimmunotherapy. The most common TRAEs of any grade were increased (n=10, 5.1%) and decreased (n=5, 2.5%) white blood cell count. When mecapegfilgrastim was administered prophylactically during the initial chemotherapy cycle, grade ≥3 neutropenia occurred in 5.1% of 178 cycles with combined immunotherapy and 6.4% of 267 cycles without immunotherapy. No FN cases were observed in the immunotherapy group. Among 119 cycles with radiotherapy, grade ≥3 neutropenia occurred in 5.0%, compared to 6.1% in the 326 cycles without radiotherapy, with one FN case reported. Across 445 chemotherapy cycles with prophylactic mecapegfilgrastim, grade ≥3 neutropenia occurred in 26 cycles (5.8%), grade 4 neutropenia in 16 cycles (3.6%), FN in 1 cycle (0.2%), and infections in 7 cycles (1.6%). Conclusions Mecapegfilgrastim demonstrated promising efficacy and safety in preventing neutropenia in patients with head and neck cancer, particularly those undergoing chemoradiotherapy or chemoimmunotherapy. Mecapegfilgrastim was well tolerated, with no new safety signals identified. These real-world findings support its continued use for neutropenia prevention in patients with head and neck cancer.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Oncology, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, China
| | - Yong Chen
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenyu Mao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ningling Zhang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Minbin Chen
- Department of Oncology, The First People’s Hospital of Kunshan, Kunshan, China
| | - Haijiao Yan
- Department of Oncology, The First People’s Hospital of Changzhou, Changzhou, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital, Jinan, China
| | - Jun Bie
- Department of Oncology, Nanchong Central Hospital, Nanchong, China
| | - Lifang Ding
- Department of Oncology, The People’s Hospital of Danyang, Danyang, China
| | - Zhanggui Wang
- Department of Radiotherapy, Anhui Provincial Second People’s Hospital, Hefei, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital, Shanghai, China
| | - Paolo Cariati
- Oral and Maxillofacial Surgery Departed, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Shukui Qin
- GI Cancer Center, Nanjing Tianyinshan Hospital, Nanjing, China
| |
Collapse
|
6
|
Ruiz-Torres DA, Bryan ME, Hirayama S, Merkin RD, Luciani E, Roberts T, Patel M, Park JC, Wirth LJ, Sadow PM, Sade-Feldman M, Stott SL, Faden DL. Immune Cell Densities Predict Response to Immune Checkpoint-Blockade in Head and Neck Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.10.24313432. [PMID: 39314968 PMCID: PMC11419212 DOI: 10.1101/2024.09.10.24313432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Immune checkpoint blockade (ICB) is the standard of care for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), yet efficacy remains low. The current approach for predicting the likelihood of response to ICB is a single proportional biomarker (PD-L1) expressed in immune and tumor cells (Combined Positive Score, CPS) without differentiation by cell type, potentially explaining its limited predictive value. Tertiary Lymphoid Structures (TLS) have shown a stronger association with ICB response than PD-L1. However, their exact composition, size, and spatial biology in HNSCC remain understudied. A detailed understanding of TLS is required for future use as a clinically applicable predictive biomarker. Methods Pre-ICB tumor tissue sections were obtained from 9 responders (complete response, partial response, or stable disease) and 11 non-responders (progressive disease) classified via RECISTv1.1. A custom multi-immunofluorescence (mIF) staining assay was designed, optimized, and applied to characterize tumor cells (pan-cytokeratin), T cells (CD4, CD8), B cells (CD19, CD20), myeloid cells (CD16, CD56, CD163), dendritic cells (LAMP3), fibroblasts (α Smooth Muscle Actin), proliferative status (Ki67) and immunoregulatory molecules (PD1). Spatial metrics were compared among groups. Serial tissue sections were scored for TLS in both H&E and mIF slides. A machine learning model was employed to measure the effect of these metrics on achieving a response to ICB (SD, PR, or CR). Results A higher density of B lymphocytes (CD20+) was found in responders compared to non-responders to ICB (p=0.022). A positive correlation was observed between mIF and pathologist identification of TLS (R 2 = 0.66, p-value= <0.0001). TLS trended toward being more prevalent in responders to ICB (p=0.0906). The presence of TLS within 100 μm of the tumor was associated with improved overall (p=0.04) and progression-free survival (p=0.03). A multivariate machine learning model identified TLS density as a leading predictor of response to ICB with 80% accuracy. Conclusion Immune cell densities and TLS spatial location within the tumor microenvironment play a critical role in the immune response to HNSCC and may potentially outperform CPS as a predictor of ICB response.
Collapse
Affiliation(s)
- Daniel A. Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts Eye and Ear, Boston, MA 02118, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
| | - Michael E. Bryan
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts Eye and Ear, Boston, MA 02118, USA
| | - Shun Hirayama
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts Eye and Ear, Boston, MA 02118, USA
| | - Ross D. Merkin
- Massachusetts Eye and Ear, Boston, MA 02118, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
| | - Evelyn Luciani
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
| | - Thomas Roberts
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
| | - Manisha Patel
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
| | - Jong C. Park
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
| | - Lori J. Wirth
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
| | - Peter M. Sadow
- Massachusetts Eye and Ear, Boston, MA 02118, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Moshe Sade-Feldman
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shannon L. Stott
- Massachusetts General Hospital Cancer Center, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel L. Faden
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts Eye and Ear, Boston, MA 02118, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
7
|
Chang SR, Chou CH, Tu HF, Liu CJ, Chang KW, Lin SC. The expression of immune co-stimulators as a prognostic predictor of head and neck squamous cell carcinomas and oral squamous cell carcinomas. J Dent Sci 2024; 19:1380-1388. [PMID: 39035328 PMCID: PMC11259670 DOI: 10.1016/j.jds.2024.05.004] [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: 04/28/2024] [Revised: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose T cells require second immune checkpoint molecules for activation and immune memory after antigen presentation. We found that inducible co-stimulator (ICOS) has been a favorable prognostic factor amongst B7 immune checkpoint co-stimulators (ICSs) families in head and neck squamous cell carcinoma (HNSCC) and oral SCC (OSCC). Materials and methods This study analyzed the expression of non-B7 tumor necrosis factor (TNF) superfamily ICSs in the Cancer Genome Atlas (TCGA) HNSCC cohort, our OSCC cohort, and TCGA pan-cancer datasets. The correlation in expression, prognosis, and immune status was assessed. Results The higher expression of CD27, CD30, CD40L, death domain 3 (DR3), and OX40, presumably on the T cell surface, defined better overall survival of HNSCC patients. Besides, CD27, CD30, CD40L, and OX40 were highly correlated with ICOS expression in tumors. CD27, CD40L, and DR3 expression are higher in HPV+ HNSCC tumors than in HPV- tumors. The combined expression level of CD27/OX40 or CD27/CD40L/OX40 enables the potent survival prediction of small, less nodal involvement, early stage, and HPV + tumor subsets. Tumors expressing high CD27, CD30, CD40L, ICOS, and OX40 exhibited enhanced immune cell infiltration. The high correlation in the expression of these ICSs was also noted in the vast majority of tumor types in TCGA datasets. Conclusion The findings of this study not only confirm the potential of the concordant stimulation of CD27, CD30, CD40L, ICOS, and OX40 as a crucial strategy in cancer immunotherapy but also inspire further exploration into the field, highlighting the promising future of cancer treatment.
Collapse
Affiliation(s)
- Shi-Rou Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Hsien Chou
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsi-Feng Tu
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, Taipei Mackay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Upadhyay R, Gogineni E, Tocaj G, Ma SJ, Bonomi M, Bhateja P, Konieczkowski DJ, Baliga S, Mitchell DL, Jhawar SR, Zhu S, Grecula JC, Dibs K, Gamez ME, Blakaj DM. Palliative Quad Shot Radiation Therapy with or without Concurrent Immune Checkpoint Inhibition for Head and Neck Cancer. Cancers (Basel) 2024; 16:1049. [PMID: 38473406 DOI: 10.3390/cancers16051049] [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: 02/20/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
OBJECTIVES Patients with recurrent and metastatic head and neck cancer (HNC) have limited treatment options. 'QuadShot' (QS), a hypofractionated palliative radiotherapy regimen, can provide symptomatic relief and local control and may potentiate the effects of immune checkpoint inhibitors (ICIs). We compared outcomes of QS ± concurrent ICIs in the palliative treatment of HNC. MATERIALS AND METHODS We identified patients who received ≥three cycles of QS from 2017 to 2022 and excluded patients without post-treatment clinical evaluation or imaging. Outcomes for patients who received QS alone were compared to those treated with ICI concurrent with QS, defined as receipt of ICI within 4 weeks of QS. RESULTS Seventy patients were included, of whom 57% received concurrent ICI. Median age was 65.5 years (interquartile range [IQR]: 57.9-77.8), and 50% patients had received prior radiation to a median dose of 66 Gy (IQR: 60-70). Median follow-up was 8.8 months. Local control was significantly higher with concurrent ICIs (12-month: 85% vs. 63%, p = 0.038). Distant control (12-month: 56% vs. 63%, p = 0.629) and median overall survival (9.0 vs. 10.0 months, p = 0.850) were similar between the two groups. On multivariable analysis, concurrent ICI was a significant predictor of local control (HR for local failure: 0.238; 95% CI: 0.073-0.778; p = 0.018). Overall, 23% patients experienced grade 3 toxicities, which was similar between the two groups. CONCLUSIONS The combination of QS with concurrent ICIs was well tolerated and significantly improved local control compared to QS alone. The median OS of 9.4 months compares favorably to historical controls for patients with HNC treated with QS. This approach represents a promising treatment option for patients with HNC unsuited for curative-intent treatment and warrants prospective evaluation.
Collapse
Affiliation(s)
- Rituraj Upadhyay
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Emile Gogineni
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Glenis Tocaj
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sung J Ma
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Marcelo Bonomi
- Department of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Priyanka Bhateja
- Department of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - David J Konieczkowski
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sujith Baliga
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Darrion L Mitchell
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sachin R Jhawar
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Simeng Zhu
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - John C Grecula
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Khaled Dibs
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mauricio E Gamez
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dukagjin M Blakaj
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Chen C, Zhang Y, Wu X, Shen J. The role of tertiary lymphoid structure and B cells in nasopharyngeal carcinoma: Based on bioinformatics and experimental verification. Transl Oncol 2024; 41:101885. [PMID: 38295746 PMCID: PMC10846412 DOI: 10.1016/j.tranon.2024.101885] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/02/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE Transcriptomic characteristics and prognosis of tertiary lymphoid structures (TLS) and infiltrating B cells in nasopharyngeal carcinoma (NPC) remain unclear. Here, NPC transcriptomic data and clinical samples were used to investigate the role of infiltrating B cells and TLS in NPC. METHODS We investigated the gene expression and infiltrating immune cells of NPC patients and further investigated the clinical relevance of B cell and TLS signatures. Transcriptional features of infiltrating B cell subsets were revealed by single-cell RNA sequencing (scRNA-seq) analysis. Immunohistochemical (IHC) and HE staining were performed to validate the clinical relevance of infiltrating B cells and TLS in NPC samples. RESULTS 27 differentially expressed immune-related genes (IRGs) associated with prognosis were identified, including B cell marker genes CD19 and CD79B. The higher B cells and TLS signature scores were associated with better outcomes and early pathological staging in 88 NPC patients. ScRNA-seq identified five distinct B cell subsets in NPC, including the BC-4 cluster associated with poor outcomes and the BC-0 cluster associated with better outcomes. EBV infection was positively associated with the formation of TLS. Furthermore, experimental results showed that the infiltration of B cells in NPC tissues was higher than that of normal tissues, and the density of TLS in an early stage of NPC was higher than that in advanced-stage TLS. CONCLUSION Our findings demonstrate the functional importance of distinct B cell subsets in the prognosis of NPC. Additionally, we confirmed that B cells and TLS may serve as prognostic biomarkers of survival for NPC patients.
Collapse
Affiliation(s)
- Chujun Chen
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yan Zhang
- Pathology Dept., The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoting Wu
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Juan Shen
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, PR China.
| |
Collapse
|
10
|
Wang Y, Lombardo E, Huang L, Avanzo M, Fanetti G, Franchin G, Zschaeck S, Weingärtner J, Belka C, Riboldi M, Kurz C, Landry G. Comparison of deep learning networks for fully automated head and neck tumor delineation on multi-centric PET/CT images. Radiat Oncol 2024; 19:3. [PMID: 38191431 PMCID: PMC10773015 DOI: 10.1186/s13014-023-02388-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
OBJECTIVES Deep learning-based auto-segmentation of head and neck cancer (HNC) tumors is expected to have better reproducibility than manual delineation. Positron emission tomography (PET) and computed tomography (CT) are commonly used in tumor segmentation. However, current methods still face challenges in handling whole-body scans where a manual selection of a bounding box may be required. Moreover, different institutions might still apply different guidelines for tumor delineation. This study aimed at exploring the auto-localization and segmentation of HNC tumors from entire PET/CT scans and investigating the transferability of trained baseline models to external real world cohorts. METHODS We employed 2D Retina Unet to find HNC tumors from whole-body PET/CT and utilized a regular Unet to segment the union of the tumor and involved lymph nodes. In comparison, 2D/3D Retina Unets were also implemented to localize and segment the same target in an end-to-end manner. The segmentation performance was evaluated via Dice similarity coefficient (DSC) and Hausdorff distance 95th percentile (HD95). Delineated PET/CT scans from the HECKTOR challenge were used to train the baseline models by 5-fold cross-validation. Another 271 delineated PET/CTs from three different institutions (MAASTRO, CRO, BERLIN) were used for external testing. Finally, facility-specific transfer learning was applied to investigate the improvement of segmentation performance against baseline models. RESULTS Encouraging localization results were observed, achieving a maximum omnidirectional tumor center difference lower than 6.8 cm for external testing. The three baseline models yielded similar averaged cross-validation (CV) results with a DSC in a range of 0.71-0.75, while the averaged CV HD95 was 8.6, 10.7 and 9.8 mm for the regular Unet, 2D and 3D Retina Unets, respectively. More than a 10% drop in DSC and a 40% increase in HD95 were observed if the baseline models were tested on the three external cohorts directly. After the facility-specific training, an improvement in external testing was observed for all models. The regular Unet had the best DSC (0.70) for the MAASTRO cohort, and the best HD95 (7.8 and 7.9 mm) in the MAASTRO and CRO cohorts. The 2D Retina Unet had the best DSC (0.76 and 0.67) for the CRO and BERLIN cohorts, and the best HD95 (12.4 mm) for the BERLIN cohort. CONCLUSION The regular Unet outperformed the other two baseline models in CV and most external testing cohorts. Facility-specific transfer learning can potentially improve HNC segmentation performance for individual institutions, where the 2D Retina Unets could achieve comparable or even better results than the regular Unet.
Collapse
Affiliation(s)
- Yiling Wang
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Elia Lombardo
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lili Huang
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Michele Avanzo
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Medical Physics, Aviano, Italy
| | - Giuseppe Fanetti
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Radiation Oncology, Aviano, Italy
| | - Giovanni Franchin
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Radiation Oncology, Aviano, Italy
| | - Sebastian Zschaeck
- Radiation Oncology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Berlin, Germany
| | - Julian Weingärtner
- Radiation Oncology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Berlin, Germany
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Marco Riboldi
- Department of Medical Physics, Ludwig-Maximilians-Universität München, Garching, Germany
| | - Christopher Kurz
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Guillaume Landry
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
11
|
Bao J, Betzler AC, Hess J, Brunner C. Exploring the dual role of B cells in solid tumors: implications for head and neck squamous cell carcinoma. Front Immunol 2023; 14:1233085. [PMID: 37868967 PMCID: PMC10586314 DOI: 10.3389/fimmu.2023.1233085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/06/2023] [Indexed: 10/24/2023] Open
Abstract
In the tumor milieu of head and neck squamous cell carcinoma (HNSCC), distinct B cell subpopulations are present, which exert either pro- or anti-tumor activities. Multiple factors, including hypoxia, cytokines, interactions with tumor cells, and other immune infiltrating lymphocytes (TILs), alter the equilibrium between the dual roles of B cells leading to cancerogenesis. Certain B cell subsets in the tumor microenvironment (TME) exhibit immunosuppressive function. These cells are known as regulatory B (Breg) cells. Breg cells suppress immune responses by secreting a series of immunosuppressive cytokines, including IL-10, IL-35, TGF-β, granzyme B, and adenosine or dampen effector TILs by intercellular contacts. Multiple Breg phenotypes have been discovered in human and mouse cancer models. However, when compartmentalized within a tertiary lymphoid structure (TLS), B cells predominantly play anti-tumor effects. A mature TLS contains a CD20+ B cell zone with several important types of B cells, including germinal-center like B cells, antibody-secreting plasma cells, and memory B cells. They kill tumor cells via antibody-dependent cytotoxicity and phagocytosis, and local complement activation effects. TLSs are also privileged sites for local T and B cell coordination and activation. Nonetheless, in some cases, TLSs may serve as a niche for hidden tumor cells and indicate a bad prognosis. Thus, TIL-B cells exhibit bidirectional immune-modulatory activity and are responsive to a variety of immunotherapies. In this review, we discuss the functional distinctions between immunosuppressive Breg cells and immunogenic effector B cells that mature within TLSs with the focus on tumors of HNSCC patients. Additionally, we review contemporary immunotherapies that aim to target TIL-B cells. For the development of innovative therapeutic approaches to complement T-cell-based immunotherapy, a full understanding of either effector B cells or Breg cells is necessary.
Collapse
Affiliation(s)
- Jiantong Bao
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
- School of Medicine, Southeast University, Nanjing, China
| | - Annika C. Betzler
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Ulm, Head & Neck Cancer Center of the Comprehensive Cancer Center Ulm, Ulm, Germany
| |
Collapse
|
12
|
Zhang J, Liu Y, Xia L, Zhen J, Gao J, Atsushi T. Constructing heterogeneous single-cell landscape and identifying microenvironment molecular characteristics of primary and lymphatic metastatic head and neck squamous cell carcinoma. Comput Biol Med 2023; 165:107459. [PMID: 37713790 DOI: 10.1016/j.compbiomed.2023.107459] [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: 07/08/2023] [Revised: 08/05/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) accounts for more than half of head and neck tumors, roughly 90%. This study focused on constructing the heterogeneous landscape using single-cell and bulk transcriptomic data to identify molecular characteristics of the microenvironment in primary and lymphatic metastatic head and neck squamous carcinomas. METHOD The study enrolled 23 HNSCC samples with scRNA-seq data and 546 HNSCC samples from TCGA. We used Monocle to sort the cells and used CellPhoneDB to explore the cell-cell interactions. Infercnv, which was used to infer cells with apparent copy number variation based on single-cell sequencing transcriptome data. We re-evaluated HNSCC bulk RNA transcriptome data to characterize the functions of different cell types in shaping the immune microenvironment of HNSCC. RESULTS We combined genealogical reconstruction, CNV inference, and cellular interactions to uncover the characteristics of distinct cell populations in different disease states, differences in cancer and immune cell lineages of differentiation trajectories, and interactions between non-immune and immune cell. PD-1 and PD-L1/PD-L2 expressed extremely rare in T cells, the immune checkpoint molecule KLRB1-CLEC2D achieved a high-level expression. We identified three microenvironment-based HNSCC subtypes associated with the prognosis of HNSCC patients. CONCLUSIONS In summary, the present study dissected the intratumoral heterogeneity and immune microenvironment of primary and metastatic HNSCC, which is crucial to reveal the mechanisms of resistance to immunotherapy in HNSCC in different disease states and is expected to assist in the further investigation of the mechanism of HNSCC cell metastasis and guide the treatment of clinical patients.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Department of Restorative Dentistry, Graduate School of Dental Medicine, Hokkaido University, Sapporo, 0608586, Japan
| | - Yunqing Liu
- Department of Restorative Dentistry, Faculty of Dental Medicine, Hokkaido University, Sapporo, 0608586, Japan
| | - Lianheng Xia
- Department of Peripheral Vascular Diseases, First Affiliated hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Jia Zhen
- Department of Peripheral Vascular Diseases, First Affiliated hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Jie Gao
- Department of Peripheral Vascular Diseases, First Affiliated hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China.
| | - Tomokiyo Atsushi
- Department of Restorative Dentistry, Faculty of Dental Medicine, Hokkaido University, Sapporo, 0608586, Japan.
| |
Collapse
|
13
|
Shi Y, Ren X, Cao S, Chen X, Yuan B, Brasil da Costa FH, Rodriguez Rosario AE, Corona A, Michikawa C, Veeramachaneni R, Osman AA, Xie T, Wang W, Sikora AG, Myers JN, Rangel R. TP53 gain-of-function mutation modulates the immunosuppressive microenvironment in non-HPV-associated oral squamous cell carcinoma. J Immunother Cancer 2023; 11:e006666. [PMID: 37604640 PMCID: PMC10445354 DOI: 10.1136/jitc-2023-006666] [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] [Accepted: 07/31/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND TP53, the most mutated gene in solid cancers, has a profound impact on most hallmarks of cancer. Somatic TP53 mutations occur in high frequencies in head and neck cancers, including oral squamous cell carcinoma (OSCC). Our study aims to understand the role of TP53 gain-of-function mutation in modulating the tumor immune microenvironment (TIME) in OSCC. METHODS Short hairpin RNA knockdown of mutant p53R172H in syngeneic oral tumors demonstrated changes in tumor growth between immunocompetent and immunodeficient mice. HTG EdgeSeq targeted messenger RNA sequencing was used to analyze cytokine and immune cell markers in tumors with inactivated mutant p53R172H. Flow cytometry and multiplex immunofluorescence (mIF) confirmed the role of mutant p53R172H in the TIME. The gene expression of patients with OSCC was analyzed by CIBERSORT and mIF was used to validate the immune landscape at the protein level. RESULTS Mutant p53R172H contributes to a cytokine transcriptome network that inhibits the infiltration of cytotoxic CD8+ T cells and promotes intratumoral recruitment of regulatory T cells and M2 macrophages. Moreover, p53R172H also regulates the spatial distribution of immunocyte populations, and their distribution between central and peripheral intratumoral locations. Interestingly, p53R172H-mutated tumors are infiltrated with CD8+ and CD4+ T cells expressing programmed cell death protein 1, and these tumors responded to immune checkpoint inhibitor and stimulator of interferon gene 1 agonist therapy. CIBERSORT analysis of human OSCC samples revealed associations between immune cell populations and the TP53R175H mutation, which paralleled the findings from our syngeneic mouse tumor model. CONCLUSIONS These findings demonstrate that syngeneic tumors bearing the TP53R172H gain-of-function mutation modulate the TIME to evade tumor immunity, leading to tumor progression and decreased survival.
Collapse
Affiliation(s)
- Yewen Shi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyong Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shaolong Cao
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xi Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bo Yuan
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Alanis E Rodriguez Rosario
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Arnoldo Corona
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chieko Michikawa
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ratna Veeramachaneni
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tongxin Xie
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenyi Wang
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew G Sikora
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roberto Rangel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
14
|
Ruffin AT, Li H, Vujanovic L, Zandberg DP, Ferris RL, Bruno TC. Improving head and neck cancer therapies by immunomodulation of the tumour microenvironment. Nat Rev Cancer 2023; 23:173-188. [PMID: 36456755 PMCID: PMC9992112 DOI: 10.1038/s41568-022-00531-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2022] [Indexed: 12/03/2022]
Abstract
Targeted immunotherapy has improved patient survival in head and neck squamous cell carcinoma (HNSCC), but less than 20% of patients produce a durable response to these treatments. Thus, new immunotherapies that consider all key players of the complex HNSCC tumour microenvironment (TME) are necessary to further enhance tumour-specific T cell responses in patients. HNSCC is an ideal tumour type in which to evaluate immune and non-immune cell differences because of two distinct TME aetiologies (human papillomavirus (HPV)-positive and HPV-negative disease), multiple anatomic sites for tumour growth, and clear distinctions between patients with locally advanced disease and those with recurrent and/or metastatic disease. Recent technological and scientific advancements have provided a more complete picture of all cellular constituents within this complex TME and have evaluated the interplay of both immune and non-immune cells within HNSCC. Here, we include a comprehensive analysis of the complete ecosystem of the HNSCC TME, performed utilizing data-rich resources such as The Cancer Genome Atlas, and cutting-edge techniques, such as single-cell RNA sequencing, high-dimensional flow cytometry and spatial multispectral imaging, to generate improved treatment strategies for this diverse disease.
Collapse
Affiliation(s)
- Ayana T Ruffin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Housaiyin Li
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Molecular Genetics and Developmental Biology (MGDB) Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lazar Vujanovic
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dan P Zandberg
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert L Ferris
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Tumour Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Taneja N, Alam A, Patnaik RS, Taneja T. Current Trends in Anticancer Drug Delivery System for Oral Cancer- A PRISMA complaint Systematic Review. Open Dent J 2022. [DOI: 10.2174/18742106-v16-e2206275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background:
Oral cancer is a deadly disease affecting worldwide. Despite developments of conventional cancer therapy, there has been little improvement in the survival rates. This culminated in the evolution of a targeted. New Drug Delivery System, discovering novel objectives for successful drug delivery and synergistic combination of anticancer agents to minimize side effects.
Objective:
The main focus was on understanding the various aspects of different targeted drug delivery vehicles used in the treatment of oral cancer including advantages, disadvantages, and future perspectives.
Materials and Methods:
A literature search was accomplished from 2005 to 2020 via Google scholar. PubMed, EBSCO, Embase, and Scopus databases along with Clinical trials registries using the terms oral buccal thin films, Hyperthermia and Thermoablation, Intra-tumoral, Photodynamic, Immunotherapy, photothermal, and ultrasound therapy in oral cancer. The articles were scrutinized and those which were not relevant to our search were omitted. Clinical trials on targeted drug delivery systems for Oral Cancer being conducted or completed around the world from various registries of clinical trials have also been searched out and the findings were tabulated in the end. The PRISMA 2020 guidelines were followed.
Results:
The treatment of oral squamous cell carcinoma (OSCC) mostly depends upon the location, type, and stage of the tumor. Vivid targeted drug delivery systems are being used in the therapeutic interventions of oral cancer as they aim for specific target site delivery and are the most appropriate treatment. Active Pharmacological Ingredient (API) is taken to the targeting site, sparing non-target organs or cells, triggering selective and efficient localization, thereby maximizing the therapeutic index with minimizing toxicity. The successful targeted drug delivery system works on four principles i.e. Retain, Evade, Target and Release, which means loading of sufficient drug into a suitable drug carrier, does not affect body secretions, long duration in circulation, reaching the targeted site and, drug release within the time for effective functioning of the drug. All techniques described in this paper have proven to show effective results.
Conclusion:
Oral Cancer is an emerging public health problem worldwide. Various conventional therapies are used for treating oral cancer, but they enclose variable degrees of side effects both on the body as well as the cellular microenvironment. With advanced technology, many other aids have been introduced in the field of oncology to treat oral cancer with minimal side effects. All techniques described in this paper have proven to show effective results in the therapeutic interventions of oral cancer. Moreover, they can be used even in combination with conventional drug therapy to show beneficial outcomes. Several clinical trials are being conducted and completed in this aspect to investigate definite results of these therapies, yet robust research is needed for further confirmation.
Collapse
|
16
|
Pereira D, Martins D, Mendes F. Immunotherapy in Head and Neck Cancer When, How, and Why? Biomedicines 2022; 10:biomedicines10092151. [PMID: 36140252 PMCID: PMC9495940 DOI: 10.3390/biomedicines10092151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/23/2022] Open
Abstract
Head and neck cancer (HNC) is one of the most common cancers worldwide. Alcohol and tobacco consumption, besides viral infections, are the main risk factors associated with this cancer. When diagnosed in advanced stages, HNC patients present a higher probability of recurrence or metastasising. The complexity of therapeutic options and post-treatment surveillance is associated with poor prognosis and reduced overall survival (OS). This review aims to explore immunotherapy (immune checkpoint inhibitors (ICI), therapeutic vaccines, and oncolytic viruses) in HNC patients’ treatment, and to explore when, how, and why patients can benefit from it. The monotherapy with ICI or in combination with chemotherapy (QT) shows the most promising results. Compared to standard therapy, ICI are able to increase OS and patients’ quality of life. QT in combination with ICI demonstrates significant response rates and considerable long-term clinical benefits. However, the toxicity associated with this approach is still a hurdle to overcome. In parallel, the therapeutic vaccines directed to the Human Papilloma Virus are also efficient in increasing the antitumour response, inducing cellular and humoral immunity. Although these results demonstrate clinical benefits compared to standard therapy, it is also important to unravel the resistance mechanisms in order to predict the clinical benefit of immunotherapy.
Collapse
Affiliation(s)
- Daniela Pereira
- Politécnico de Coimbra, ESTeSC, UCPCBL, Rua 5 de Outubro–SM Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
| | - Diana Martins
- Politécnico de Coimbra, ESTeSC, UCPCBL, Rua 5 de Outubro–SM Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
- Laboratório de Investigação em Ciências Aplicadas à Saúde (LabinSaúde), Politécnico de Coimbra, ESTESC, Rua 5 de Outubro–SM Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Fernando Mendes
- Politécnico de Coimbra, ESTeSC, UCPCBL, Rua 5 de Outubro–SM Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
- Laboratório de Investigação em Ciências Aplicadas à Saúde (LabinSaúde), Politécnico de Coimbra, ESTESC, Rua 5 de Outubro–SM Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- European Association for Professions in Biomedical Sciences, B-1000 Brussels, Belgium
- Correspondence:
| |
Collapse
|
17
|
Starzer AM, Preusser M, Berghoff AS. Immune escape mechanisms and therapeutic approaches in cancer: the cancer-immunity cycle. Ther Adv Med Oncol 2022; 14:17588359221096219. [PMID: 35510032 PMCID: PMC9058458 DOI: 10.1177/17588359221096219] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/04/2022] [Indexed: 12/31/2022] Open
Abstract
The introduction of immune checkpoint inhibitors has changed the therapeutic possibilities for various cancer types. However, despite the success in some entities, a significant fraction of patients does not respond to immune checkpoint inhibitors. A functioning cancer-immunity cycle is needed as the precondition for a clinically meaningful response to immune checkpoint inhibitors. It is assumed that only if each step of the cycle is activated and functioning properly, immune checkpoint inhibitors induce a meaningful immune response. However, an activated cancer-immunity cycle might not be present equally in each patient and cancer type. Ideally, treatment concepts should consider each single step of the cancer-immunity cycle and provide personalized treatment approaches, allowing the adaption to functioning and malfunctioning steps of the individual patient’s specific cancer-immunity cycle. In the following review, we provide an overview of the single steps of the cancer-immunity cycle as well as the impact of malfunctioning steps on the generation of an effective tumor-specific immune response.
Collapse
Affiliation(s)
- Angelika M. Starzer
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S. Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
de Sousa LG, Ferrarotto R. Pembrolizumab in the first-line treatment of advanced head and neck cancer. Expert Rev Anticancer Ther 2021; 21:1321-1331. [PMID: 34689660 DOI: 10.1080/14737140.2021.1996228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Recurrent or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) is associated with dismal prognosis and has limited therapeutic options. PD-1/PD-L1 axis blockade was initially shown to improve outcomes in platinum-refractory HNSCC. More recently, pembrolizumab monotherapy or pembrolizumab combined with chemotherapy resulted in better overall survival than platinum, 5-fluorouracil, and cetuximab (EXTREME regimen) as first-line therapy for R/M HNSCC, establishing a new standard-of-care therapy for this disease. AREAS COVERED We review pembrolizumab in the first-line treatment of R/M HNSCC and summarize the impact of PD-L1 expression, tumor and symptom burden, and patient's performance status on treatment decisions. Future perspectives are summarized. EXPERT OPINION The standard-of-care first-line therapy for R/M HNSCC is pembrolizumab monotherapy for patients with a PD-L1 combined positive score (CPS)≥1 or pembrolizumab combined with platinum and 5-fluorouracil for patients with any PD-L1 status. Addition of chemotherapy to pembrolizumab increases the response rate but also toxicity and is preferred for patients with good performance status and significant tumor and symptom burden. For patients with a PD-L1 CPS <1, the EXTREME regimen should be considered. New strategies combining pembrolizumab with targeted therapies and immune checkpoints inhibitors are being explored to synergize or overcome resistance to anti-PD-1.
Collapse
Affiliation(s)
- Luana Guimaraes de Sousa
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas Md Anderson Cancer Center, Houston, Texas, USA
| | - Renata Ferrarotto
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas Md Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
19
|
Ruffin AT, Cillo AR, Tabib T, Liu A, Onkar S, Kunning SR, Lampenfeld C, Atiya HI, Abecassis I, Kürten CHL, Qi Z, Soose R, Duvvuri U, Kim S, Oesterrich S, Lafyatis R, Coffman LG, Ferris RL, Vignali DAA, Bruno TC. B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma. Nat Commun 2021; 12:3349. [PMID: 34099645 PMCID: PMC8184766 DOI: 10.1038/s41467-021-23355-x] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 04/21/2021] [Indexed: 01/06/2023] Open
Abstract
Current immunotherapy paradigms aim to reinvigorate CD8+ T cells, but the contribution of humoral immunity to antitumor immunity remains understudied. Here, we demonstrate that in head and neck squamous cell carcinoma (HNSCC) caused by human papillomavirus infection (HPV+), patients have transcriptional signatures of germinal center (GC) tumor infiltrating B cells (TIL-Bs) and spatial organization of immune cells consistent with tertiary lymphoid structures (TLS) with GCs, both of which correlate with favorable outcome. GC TIL-Bs in HPV+ HNSCC are characterized by distinct waves of gene expression consistent with dark zone, light zone and a transitional state of GC B cells. Semaphorin 4a expression is enhanced on GC TIL-Bs present in TLS of HPV+ HNSCC and during the differentiation of TIL-Bs. Our study suggests that therapeutics to enhance TIL-B responses in HNSCC should be prioritized in future studies to determine if they can complement current T cell mediated immunotherapies.
Collapse
Affiliation(s)
- Ayana T Ruffin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | - Tracy Tabib
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angen Liu
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sayali Onkar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sheryl R Kunning
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | - Caleb Lampenfeld
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | - Huda I Atiya
- Hillman Cancer Center, Pittsburgh, PA, USA
- Division of Hematology and Oncology, Department of Medicine, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Irina Abecassis
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Zengbiao Qi
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan Soose
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Umamaheswar Duvvuri
- Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seungwon Kim
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steffi Oesterrich
- Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Women's Cancer Research Center, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lan G Coffman
- Hillman Cancer Center, Pittsburgh, PA, USA
- Division of Hematology and Oncology, Department of Medicine, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert L Ferris
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Hillman Cancer Center, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
20
|
Peng Y, Xiao L, Rong H, Ou Z, Cai T, Liu N, Li B, Zhang L, Wu F, Lan T, Lin X, Li Q, Ren S, Fan S, Li J. Single-cell profiling of tumor-infiltrating TCF1/TCF7 + T cells reveals a T lymphocyte subset associated with tertiary lymphoid structures/organs and a superior prognosis in oral cancer. Oral Oncol 2021; 119:105348. [PMID: 34044317 DOI: 10.1016/j.oraloncology.2021.105348] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Despite substantial advances in treatment, clinical outcomes for oral squamous cell carcinoma (OSCC) remain unsatisfactory. Tumor-infiltrating lymphocytes (TILs) are an important prognostic factor for patients and are heterogeneous. Some studies have suggested that TCF1/TCF7+ T cells and tertiary lymphatic structure/organ (TLS) play an important role in tumor immunity. However, how they affect tumor immunity and whether they are related to prognosis in OSCC have not been reported in detail. MATERIALS AND METHODS We isolated OSCC cells and performed single-cell RNA sequencing (scRNA-seq). We used immunohistochemistry (IHC) to analyze the relationship between TLSs and prognosis. Multiplex immunohistochemistry (MIHC), flow cytometry (FCM) and spatial analysis were performed to verify the characteristics of TCF1/TCF7+ T cells. The prognostic significance and upstream regulatory network of the TCF1/TCF7+ T cell subpopulation were determined by multivariate analysis and Scenic software. RESULTS We found a strong association between TCF1/TCF7+ T cell subsets, TLSs and prognosis. The results suggested that TCF1/TCF7+ T cells express high levels of TLS-related genes and low levels of immune checkpoint molecules. Finally, we found that TCF1/TCF7+ T cells were significantly associated with favorable outcomes. We also describe the upstream drivers that these cells rely on. CONCLUSIONS TCF1/TCF7+ T cells could be used as a new therapeutic target to regulate the immune response of OSCC and are expected to be a new prognostic marker.
Collapse
Affiliation(s)
- Yu Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Liping Xiao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Haixu Rong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Zhanpeng Ou
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Tingting Cai
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Niu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Bowen Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Lizao Zhang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Fan Wu
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Tianjun Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China
| | - Xinyu Lin
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Qunxing Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Siqi Ren
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China.
| | - Jinsong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-sen Memorial Hospital, Guangzhou 510120, China; Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
21
|
Shi Y, Xie TX, Leach DG, Wang B, Young S, Osman AA, Sikora AG, Ren X, Hartgerink JD, Myers JN, Rangel R. Local Anti-PD-1 Delivery Prevents Progression of Premalignant Lesions in a 4NQO-Oral Carcinogenesis Mouse Model. Cancer Prev Res (Phila) 2021; 14:767-778. [PMID: 34021022 DOI: 10.1158/1940-6207.capr-20-0607] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/21/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Although the principle of systemic treatment to prevent the progression of oral premalignant lesions (OPL) has been demonstrated, there remains a lack of consensus about an optimal approach that balances clinical efficacy with toxicity concerns. Recent advances in cancer therapy using approaches targeting the tumor immune microenvironment (TIME) including immune-checkpoint inhibitors indicate that these agents have significant clinically activity against different types of cancers, including oral cancer, and therefore they may provide an effective oral cancer prevention strategy for patients with OPLs. Our past work showed that systemic delivery of a monoclonal antibody to the programmed death receptor 1 (PD-1) immune checkpoint can inhibit the progression of OPLs to oral cancer in a syngeneic murine oral carcinogenesis model. Here we report a novel approach of local delivery of a PD-1 immune-checkpoint inhibitor loaded using a hydrogel, which significantly reduces the progression of OPLs to carcinomas. In addition, we detected a significant infiltration of regulatory T cells associated with oral lesions with p53 mutation, and a severe loss of expression of STING, which correlated with a decreased infiltration of dendritic cells in the oral lesions. However, a single local dose of PD-1 inhibitor was found to restore stimulator of interferon response cGAMP interactor 1 (STING) and CD11c expression and increase the infiltration of CD8+ T cells into the TIME irrespective of the p53 mutational status. Overall, we provide evidence for the potential clinical value of local delivery of biomaterials loaded with anti-PD-1 antibodies to prevent malignant progression of OPLs. PREVENTION RELEVANCE: Oral cancer is an aggressive disease, with an overall survival rate of 50%. Preinvasive histologic abnormalities such as tongue dysplasia represent an early stage of oral cancer; however, there are no treatments to prevent oral carcinoma progression. Here, we combined biomaterials loaded with an immunotherapeutic agent preventing oral cancer progression.
Collapse
Affiliation(s)
- Yewen Shi
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tong-Xin Xie
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David G Leach
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, Texas
| | - Bingbing Wang
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simon Young
- Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Abdullah A Osman
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew G Sikora
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoyong Ren
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jeffrey D Hartgerink
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, Texas
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roberto Rangel
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
22
|
Zhang X, Yang J. Role of Non-coding RNAs on the Radiotherapy Sensitivity and Resistance of Head and Neck Cancer: From Basic Research to Clinical Application. Front Cell Dev Biol 2021; 8:637435. [PMID: 33644038 PMCID: PMC7905100 DOI: 10.3389/fcell.2020.637435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Head and neck cancers (HNCs) rank as the sixth common and the seventh leading cause of cancer-related death worldwide, with an estimated incidence of 600,000 cases and 40-50% mortality rate every year. Radiotherapy is a common local therapeutic modality for HNC mainly through the function of ionizing radiation, with approximately 60% of patients treated with radiotherapy or chemoradiotherapy. Although radiotherapy is more advanced and widely used in clinical practice, the 5-year overall survival rates of locally advanced HNCs are still less than 40%. HNC cell resistance to radiotherapy remains one of the major challenges to improve the overall survival in HNC patients. Non-coding RNAs (ncRNAs) are newly discovered functional small RNA molecules that are different from messenger RNAs, which can be translated into a protein. Many previous studies have reported the dysregulation and function of ncRNAs in HNC. Importantly, researchers reported that several ncRNAs were also dysregulated in radiotherapy-sensitive or radiotherapy-resistant HNC tissues compared with the normal cancer tissues. They found that ectopically elevating or knocking down expression of some ncRNAs could significantly influence the response of HNC cancer cells to radiotherapy, indicating that ncRNAs could regulate the sensitivity of cancer cells to radiotherapy. The implying mechanism for ncRNAs in regulating radiotherapy sensitivity may be due to its roles on affecting DNA damage sensation, inducing cell cycle arrest, regulating DNA damage repair, modulating cell apoptosis, etc. Additionally, clinical studies reported that in situ ncRNA expression in HNC tissues may predict the response of radiotherapy, and circulating ncRNA from body liquid serves as minimally invasive therapy-responsive and prognostic biomarkers in HNC. In this review, we aimed to summarize the current function and mechanism of ncRNAs in regulating the sensitivity of HNC cancer cells to radiotherapy and comprehensively described the state of the art on the role of ncRNAs in the prognosis prediction, therapy monitoring, and prediction of response to radiotherapy in HNC.
Collapse
Affiliation(s)
- Xixia Zhang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
23
|
Green SE, McCusker MG, Mehra R. Emerging immune checkpoint inhibitors for the treatment of head and neck cancers. Expert Opin Emerg Drugs 2020; 25:501-514. [PMID: 33196319 DOI: 10.1080/14728214.2020.1852215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: The benefits of immune checkpoint inhibitors (ICIs) in recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) have been demonstrated through multiple studies to improve overall survival (OS) with decreased side effects when compared to the standard of care (SOC) treatment regimens in place for decades, leading to the approval of two ICIs, nivolumab and pembrolizumab. There has been a subsequent influx in the development of novel immunotherapy agents for the treatment of HNSCC. Areas covered: Data for anti-programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4) antibodies in treatment of R/M HNSCC will be reviewed. Emerging immune checkpoint inhibitors as well as combined therapies in HNSCC will be discussed. The role of predictive biomarkers, HPV-status, PD-L1 expression, and challenges related to treating patients with ICIs will be summarized. Expert opinion: A shift toward ICIs as SOC for the treatment of R/M HNSCC will continue as emerging immune checkpoints and combination therapies are evaluated. Response rates are variable in this patient population underlying the importance of identifying predictive biomarkers to aid in patient selection for ICI treatment.
Collapse
Affiliation(s)
- Sarah E Green
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| | - Michael G McCusker
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| | - Ranee Mehra
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| |
Collapse
|
24
|
Qiu Y, Cui L, Lin Y, Gao B, Li J, Zhao X, Zhu X, Hu S, Lin L. Development and Validation of a Robust Immune Prognostic Signature for Head and Neck Squamous Cell Carcinoma. Front Oncol 2020; 10:1502. [PMID: 33224866 PMCID: PMC7667274 DOI: 10.3389/fonc.2020.01502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is among the most destructive of tumors, leading to considerable morbidity and mortality. Abnormal immune microenvironment is closely associated with tumor progression. This study aimed to construct a robust immune prognostic model for HNSCC. The RNA-seq transcriptome data and clinical information of HNSCC were downloaded from The Cancer Genome Atlas (TCGA) database. The key pathways and transcriptional factors (TFs) that are correlated with significantly altered immune related genes were identified. A robust immune prognostic model was constructed and further validated using a discovery-validation cohort design. An immune prognostic signature-based nomogram model was also developed. We have identified 400 significantly changed immune related genes in HNSCC. In addition, functional analysis of the altered immune related genes revealed many biological functions and pathways that might affect the tumor immune microenvironment. FOXP3, SNAI2, and STAT1 were identified as the hub TFs for regulating immunological changes in HNSCC. Moreover, an immune related gene-based prognostic signature significantly associated with the overall survival (OS) of HNSCC was constructed in the discovery cohort, and successfully validated in the validation cohort. Finally, a nomogram model based on immune prognostic signature was built and exhibited good performance for predicting the OS of HNSCC. In conclusion, the immune prognostic model is robust for predicting the prognosis of HNSCC and may evolve as a promising tool for risk evaluation and therapeutic selection.
Collapse
Affiliation(s)
- Yu Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| | - Li Cui
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yang Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| | - Bingju Gao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| | - Jun Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| | - Xinyuan Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofeng Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| | - Shen Hu
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lisong Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Maxillofacial Medicine Center of Fujian Province, Fuzhou, China
| |
Collapse
|
25
|
Plavc G, Strojan P. Combining radiotherapy and immunotherapy in definitive treatment of head and neck squamous cell carcinoma: review of current clinical trials. Radiol Oncol 2020; 54:377-393. [PMID: 33064670 PMCID: PMC7585335 DOI: 10.2478/raon-2020-0060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) presents as locally advanced disease in a majority of patients and is prone to relapse despite aggressive treatment. Since immune checkpoint inhibitors (ICI) have shown clinically significant efficacy in patients with recurrent/metastatic HNSCC (R/M HNSCC), a plethora of trials are investigating their role in earlier stages of disease. At the same time, preclinical data showed the synergistic role of concurrently administered radiotherapy and ICIs (immunoradiotherapy) and explained several mechanisms behind it. Therefore, this approach is prospectively tested in a neoadjuvant, definitive, or adjuvant setting in non-R/M HNSCC patients. Due to the intricate relationship between host, immunotherapy, chemotherapy, and radiotherapy, each of these approaches has its advantages and disadvantages. In this narrative review we present the biological background of immunoradiotherapy, as well as a rationale for, and possible flaws of, each treatment approach, and provide readers with a critical summary of completed and ongoing trials. Conclusions While immunotherapy with ICIs has already become a standard part of treatment in patients with R/M HNSCC, its efficacy in a non-R/M HNSCC setting is still the subject of extensive clinical testing. Irradiation can overcome some of the cancer's immune evasive manoeuvres and can lead to a synergistic effect with ICIs, with possible additional benefits of concurrent platinum-based chemotherapy. However, the efficacy of this combination is not robust and details in trial design and treatment delivery seem to be of unprecedented importance.
Collapse
Affiliation(s)
- Gaber Plavc
- Department of Radiation Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Primoz Strojan
- Department of Radiation Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
26
|
Borel C, Jung AC, Burgy M. Immunotherapy Breakthroughs in the Treatment of Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:E2691. [PMID: 32967162 PMCID: PMC7563963 DOI: 10.3390/cancers12092691] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) in the recurrent or metastatic (R/M) setting is a devastating disease with a poor prognosis. Until recently, the reference first line treatment was the EXTREME protocol, which yields a 10.1 months median survival, and almost no effective treatment are available in second line. Immune checkpoint inhibitors (ICIs) have changed the prognosis of several metastatic solid tumors. Given their inflammatory profile and high mutational burden, HNSCC is a good candidate for ICIs treatments. First, a strong pembrolizumab efficacy signal was shown in the Keynote-012 Phase Ib study. Then, the phase III Checkmate-141 study validated the efficacy of nivolumab in platinum-resistant patients. Finally, the first line conquest is acquired since the final results of the keynote-048 phase III study that demonstrated the superiority of pembrolizumab versus EXTREME in CPS ≥ 1 patients, and with the addition of platinum and 5FU in all patients. However, the first line treatment landscape is not frozen. Two studies (Checkmate-651 and Kestrel) are investigating the efficacy of the combination of antibodies raised against CTLA-4 and PD-(L)1. Results are impatiently awaited. Further progress needs the use of new immunotherapeutic agents such as monalizumab or ICOS agonist rather in combination with an anti-PD(L)1. New associations of ICIs and chemotherapeutic or targeted therapeutic agents are also actively investigated. Finally, ICIs has to be studied in the locally advanced setting where there is a chance of cure. Several trials are testing the potential synergistic combination of ICIs with radiotherapy and platinum or cetuximab, or ICIs used in a neoadjuvant setting.
Collapse
Affiliation(s)
- Christian Borel
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France;
| | - Alain C. Jung
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France;
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France
| | - Mickaël Burgy
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France;
| |
Collapse
|
27
|
Mazloom A, Ghalehsari N, Gazivoda V, Nimkar N, Paul S, Gregos P, Rateshwar J, Khan U. Role of Immune Checkpoint Inhibitors in Gastrointestinal Malignancies. J Clin Med 2020; 9:E2533. [PMID: 32781500 PMCID: PMC7463795 DOI: 10.3390/jcm9082533] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of several solid and hematological malignancies. ICIs are not only able to produce long and durable responses, but also very well tolerated by patients. There are several approved indications of use of ICIs in treatment of metastatic gastrointestinal malignancies including gastric, esophageal, colorectal and hepatocellular carcinoma. In addition, ICIs can be used in microsatellite instability-high (MSI-H) and high tumor mutational burden (TMB) tumors in chemotherapy-resistant setting. Despite having good efficacy and superior safety profile, ICIs are clinically active in small subset of patients, therefore, there is a huge unmet need to enhance their efficacy and discover new predictive biomarkers. There are several ongoing clinical trials that are exploring the role of ICIs in various gastrointestinal cancers either as single agent or in combination with chemotherapy, radiation therapy, targeted agents or other immunotherapeutic agents. In this review, we discuss the published and ongoing trials for ICIs in gastrointestinal malignancies, including esophageal, gastric cancer, pancreatic, hepatocellular, biliary tract, colorectal and anal cancers. Specifically, we focus on the use of ICIs in each line of therapy and discuss the future directions of these agents in each type of gastrointestinal cancer.
Collapse
Affiliation(s)
- Anita Mazloom
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Nima Ghalehsari
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Victor Gazivoda
- Department of Surgery, Maimonides Medical Center, Brooklyn, NY 11219, USA;
| | - Neil Nimkar
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Sonal Paul
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Peter Gregos
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Janice Rateshwar
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| | - Uqba Khan
- Department of Medicine, NewYork-Presbyterian Brooklyn Methodist Hospital—Weill Cornell Medicine, Brooklyn, NY 11215, USA; (A.M.); (N.G.); (N.N.); (S.P.); (P.G.); (J.R.)
| |
Collapse
|
28
|
Suzuki S, Ogawa T, Sano R, Takahara T, Inukai D, Akira S, Tsuchida H, Yoshikawa K, Ueda R, Tsuzuki T. Immune-checkpoint molecules on regulatory T-cells as a potential therapeutic target in head and neck squamous cell cancers. Cancer Sci 2020; 111:1943-1957. [PMID: 32304268 PMCID: PMC7293074 DOI: 10.1111/cas.14422] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/26/2020] [Accepted: 04/05/2020] [Indexed: 12/18/2022] Open
Abstract
Immune-checkpoint inhibitors improve the survival of head and neck squamous cell carcinoma (HNSCC) patients. Although recent studies have demonstrated that the tumor immune microenvironment (TIME) has critical roles in immunotherapy, the precise mechanisms involved are unclear. Therefore, further investigations of TIME are required for the improvement of immunotherapy. The frequency of effector regulatory T-cells (eTregs) and the expression of immune-checkpoint molecules (ICM) on eTregs and conventional T-cells (Tconvs) both in peripheral blood lymphocytes (PBL) and tumor-infiltrating lymphocytes (TIL) from HNSCC patients were analyzed by flow cytometry and their distributions were evaluated by multi-color immunofluorescence microscopy. High frequency eTreg infiltration into HNSCC tissues was observed and high expressions of CD25, FOXP3, stimulatory-ICM (4-1BB, ICOS, OX40 and GITR) and inhibitory-ICM (programmed cell death-1 [PD-1] and cytotoxic T-lymphocyte-associated protein-4 [CTLA-4]) were found on invasive eTregs. In contrast, the expression of stimulatory-ICM on Tconvs was low and the expression of inhibitory-ICM was high. In addition, ICM-ligands (programmed cell death-1 [PD-L1], galectin-9 and CEACAM-1) were frequently expressed on cancer cells. PD-L1 and galectin-9 were also expressed on macrophages. PD-1+ T-cells interacted with PD-L1+ cancer cells or PD-L1+ macrophages. This suggested that in TIL, eTregs are highly activated, but Tconvs are exhausted or inactivated by eTregs and immune-checkpoint systems, and ICM and eTregs are strongly involved in the creation of an immunosuppressive environment in HNSCC tissues. These suggested eTreg targeting drugs are expected to be a combination partner with immune-checkpoint inhibitors that will improve immunotherapy of HNSCC.
Collapse
Affiliation(s)
- Susumu Suzuki
- Research Creation Support Centre, Aichi Medical University, Nagakute, Japan.,Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Tetsuya Ogawa
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Rui Sano
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Taishi Takahara
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Japan
| | - Daisuke Inukai
- Department of Otorhinolaryngology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Satou Akira
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Japan
| | - Hiromi Tsuchida
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kazuhiro Yoshikawa
- Research Creation Support Centre, Aichi Medical University, Nagakute, Japan
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Japan
| |
Collapse
|
29
|
Bisheshar SK, De Ruiter EJ, Devriese LA, Willems SM. The prognostic role of NK cells and their ligands in squamous cell carcinoma of the head and neck: a systematic review and meta-analysis. Oncoimmunology 2020; 9:1747345. [PMID: 32363116 PMCID: PMC7185215 DOI: 10.1080/2162402x.2020.1747345] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/21/2020] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background : Despite the improvement in therapeutic interventions, 5-year survival rates in Head and Neck Squamous Cell Carcinoma (HNSCC) are limited. HNSCC is an immunogenic cancer type for which molecular stratification markers are lacking. Tumor-infiltrating lymphocytes (TILs) have shown a favorable prognostic role in different cancer types. This study focused on the prognostic role of NK cells in HNSCC. Methods : A systematic search was conducted in Pubmed/Medline and Embase. Articles that correlated the presence of intratumoral NK cells, activating/inhibiting receptors, death receptors, or their ligands with clinicopathologic characteristics or survival were included. A meta-analysis was performed that assessed the association between CD56+ and CD57+ and overall survival (OS), disease-free survival (DFS), and progression-free survival (PFS). Results : A pooled analysis indicated a favorable prognostic role of CD56+ and CD57+ NK cells for OS (HR 0.19 CI 0.11-0.35). NK cell markers NKp46 and Granzyme B (GrB) also have a favorable prognostic role. NK cell ligand Fas correlated with better survival and better characteristics. NK cell marker Fas-L, NK cell ligands CEACAM1, RCAS1, CD70 and TRAIL-R, and effector molecules of these ligands, FADD and FAP1, correlated to features of worse prognosis. Conclusion : A favorable prognostic role of NK cells in HNSCC was found in this review. Some studies implied the opposite, indicating the fine balance between pro- and anti-tumor functions of NK cells. Future studies using homogeneous patient cohorts regarding tumor subsite and treatment modality, are necessary to further provide insight into the prognostic role of NK cells.
Collapse
Affiliation(s)
- Sangeeta K. Bisheshar
- Department of Pathology, University Medical Center Utrecht, CX Utrecht 3584, The Netherlands
| | - Emma J. De Ruiter
- Department of Pathology, University Medical Center Utrecht, CX Utrecht 3584, The Netherlands
| | - Lot A. Devriese
- Department of Medical Oncology, University Medical Center Utrecht, CX Utrecht 3584, The Netherlands
| | - Stefan M. Willems
- Department of Pathology, University Medical Center Utrecht, CX Utrecht 3584, The Netherlands
| |
Collapse
|
30
|
Li L, Wang XL, Lei Q, Sun CZ, Xi Y, Chen R, He YW. Comprehensive immunogenomic landscape analysis of prognosis-related genes in head and neck cancer. Sci Rep 2020; 10:6395. [PMID: 32286381 PMCID: PMC7156482 DOI: 10.1038/s41598-020-63148-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancer is the sixth most common malignancy around the world, and 90% of cases are squamous cell carcinomas. In this study, we performed a systematic investigation of the immunogenomic landscape to identify prognostic biomarkers for head and neck squamous cell carcinoma (HNSCC). We analyzed the expression profiles of immune-related genes (IRGs) and clinical characteristics by interrogating RNA-seq data from 527 HNSCC patients in the cancer genome atlas (TCGA) dataset, including 41 HPV+ and 486 HPV- samples. We found that differentially expressed immune genes were closely associated with patient prognosis in HNSCC by comparing the differences in gene expression between cancer and normal samples and performing survival analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to annotate the biological functions of the differentially expressed immunogenomic prognosis-related genes. Two additional cohorts from the Oncomine database were used for validation. 65, 56 differentially expressed IRGs was associated with clinical prognosis in total and HPV- samples, respectively. Furthermore, we extracted 10, 11 prognosis-related IRGs from 65, 56 differentially expressed IRGs, respectively. They were significantly correlated with clinical prognosis and used to construct the prognosis prediction models. The multivariable ROC curves (specifically, the AUC) were used to measure the accuracy of the prognostic models. These genes were mainly enriched in several gene ontology (GO) terms related to immunocyte migration and receptor and ligand activity. KEGG pathway analysis revealed enrichment of pathways related to cytokine-cytokine receptor interactions, which are primarily involved in biological processes. In addition, we identified 63 differentially expressed transcription factors (TFs) from 4784 differentially expressed genes, and 16 edges involving 18 nodes were formed in the regulatory network between differentially expressed TFs and the high-risk survival-associated IRGs. B cell and CD4 T cell infiltration levels were significantly negatively correlated with the expression of prognosis-related immune genes regardless of HPV status. In conclusion, this comprehensive analysis identified the prognostic IRGs as potential biomarkers, and the model generated in this study may enable an accurate prediction of survival.
Collapse
Affiliation(s)
- Lei Li
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Xiao-Li Wang
- Radiation Therapy Center, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Qian Lei
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Chuan-Zheng Sun
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Yan Xi
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Ran Chen
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Yong-Wen He
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Yunnan, China.
| |
Collapse
|
31
|
Clayton SM, Archard JA, Wagner J, Farwell DG, Bewley AF, Beliveau A, Birkeland A, Rao S, Abouyared M, Belafsky PC, Anderson JD. Immunoregulatory Potential of Exosomes Derived from Cancer Stem Cells. Stem Cells Dev 2020; 29:327-335. [PMID: 31856674 PMCID: PMC7081244 DOI: 10.1089/scd.2019.0197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are malignancies that originate in the mucosal lining of the upper aerodigestive tract. Despite advances in therapeutic interventions, survival rates among HNSCC patients have remained static for years. Cancer stem cells (CSCs) are tumor-initiating cells that are highly resistant to treatment, and are hypothesized to contribute to a significant fraction of tumor recurrences. Consequently, further investigations of how CSCs mediate recurrence may provide insights into novel druggable targets. A key element of recurrence involves the tumor's ability to evade immunosurveillance. Recent published reports suggest that CSCs possess immunosuppressive properties, however, the underlying mechanism have yet to be fully elucidated. To date, most groups have focused on the role of CSC-derived secretory proteins, such as cytokines and growth factors. Here, we review the established immunoregulatory role of exosomes derived from mixed tumor cell populations, and propose further study of CSC-derived exosomes may be warranted. Such studies may yield novel insights into new druggable targets, or lay the foundation for future exosome-based diagnostics.
Collapse
Affiliation(s)
- Shannon M. Clayton
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Joehleen A. Archard
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Joseph Wagner
- University of California Drug Discovery Consortium, University of California, Davis, Sacramento, California
| | - D. Gregory Farwell
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Arnaud F. Bewley
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Angela Beliveau
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Andrew Birkeland
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Shyam Rao
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Marianne Abouyared
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Peter C. Belafsky
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| | - Johnathon D. Anderson
- Department of Otolaryngology, University of California, Davis, Sacramento, California
| |
Collapse
|
32
|
Overexpression of LAPTM4B-35 is a negative prognostic factor in head and neck squamous cell carcinoma. Sci Rep 2019; 9:18866. [PMID: 31827181 PMCID: PMC6906383 DOI: 10.1038/s41598-019-55319-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/26/2019] [Indexed: 11/21/2022] Open
Abstract
Overexpression of LAPTM4B-35 (lysosomal-associated transmembrane protein 4β-35) is associated with a poor prognosis in numerous malignant tumours. Expression patterns and effects of LAPTM4B-35 on head and neck squamous cell carcinomas (HNSCC) are unknown. The aim of this study was to investigate the prognostic relevance of LAPTM4B-35 in HNSCC. Tissue microarrays were constructed with primary tumours and associated lymph node metastases isolated from 127 patients. The expression of LAPTM4B-35 was investigated by immunohistochemistry and the results were correlated with survival data. LAPTM4B-35 in the primary tumour was highly expressed in 47.2% of the patients (60/127). LAPTM4B-35 expression was significantly associated with tumour stage. Moreover, overexpression of LAPTM4B-35 correlated with a significantly worse disease-free survival (10.23 years vs. not reached) and a higher recurrence rate (40.7% vs. 25%). High expression of LAPTM4B-35 in lymph node metastasis was found in 29.2% of cases. In 19.4% of cases, high LAPTM4B-35 expression was observed in both the primary tumour and corresponding lymph node metastases. In conclusion, our data indicates that overexpression of LAPTM4B-35 is associated with poor prognosis and may therefore serve as a new prognostic marker in HNSCC.
Collapse
|
33
|
Doumas S, Foukas PG, Economopoulou P, Kotsantis I, Psyrri A. Atypical patterns of responses in the era of immune checkpoint inhibitors in head and neck cancer. Oral Oncol 2019; 100:104477. [PMID: 31837533 DOI: 10.1016/j.oraloncology.2019.104477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/03/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
The discovery and implementation into everyday clinical practice of immune checkpoint inhibitors (ICIs) has marked a therapeutic renaissance in the treatment of advanced solid tumors. In head and neck cancer, nivolumab and pembrolizumab have both been approved for recurrent/metastatic disease based on robust clinical activity observed in landmark phase III clinical trials. Despite tremendous improvements in overall survival, patterns of response and progression to ICIs may be distinct from those traditionally described with classical chemotherapy or molecularly targeted therapies. In this context, pseudoprogression is observed in patients treated with ICIs that show response after a transient increase in tumor burden and hyperprogression is described as rapid radiological or clinical progression after immunotherapy. Most importantly, the assessment of radiological response in patients receiving ICIs needs to be differentiated. In this review, we aim to describe radiologic criteria for immune response evaluation and illustrate the newly reported concepts of atypical patterns of response to ICIs.
Collapse
Affiliation(s)
- Stergios Doumas
- Maxillofacial Unit, Brighton & Sussex University Hospitals NHS Trust, United Kingdom
| | - Periklis G Foukas
- 2nd Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1st Rimini St, 12462 Haidari, Athens, Greece
| | - Panagiota Economopoulou
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital National and Kapodistrian University of Athens, 1st Rimini St, 12462 Haidari, Athens, Greece
| | - Ioannis Kotsantis
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital National and Kapodistrian University of Athens, 1st Rimini St, 12462 Haidari, Athens, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Second Department of Internal Medicine, Attikon University Hospital National and Kapodistrian University of Athens, 1st Rimini St, 12462 Haidari, Athens, Greece.
| |
Collapse
|
34
|
Choonoo G, Blucher AS, Higgins S, Boardman M, Jeng S, Zheng C, Jacobs J, Anderson A, Chamberlin S, Evans N, Vigoda M, Cordier B, Tyner JW, Kulesz-Martin M, McWeeney SK, Laderas T. Illuminating biological pathways for drug targeting in head and neck squamous cell carcinoma. PLoS One 2019; 14:e0223639. [PMID: 31596908 PMCID: PMC6785123 DOI: 10.1371/journal.pone.0223639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/25/2019] [Indexed: 11/18/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a morbid disease with poor prognosis and treatment that typically leaves patients with permanent damage to critical functions such as eating and talking. Currently only three targeted therapies are FDA approved for use in HNSCC, two of which are recently approved immunotherapies. In this work, we identify biological pathways involved with this disease that could potentially be targeted by current FDA approved cancer drugs and thereby expand the pool of potential therapies for use in HNSCC treatment. We analyzed 508 HNSCC patients with sequencing information from the Genomic Data Commons (GDC) database and assessed which biological pathways were significantly enriched for somatic mutations or copy number alterations. We then further classified pathways as either “light” or “dark” to the current reach of FDA-approved cancer drugs using the Cancer Targetome, a compendium of drug-target information. Light pathways are statistically enriched with somatic mutations (or copy number alterations) and contain one or more targets of current FDA-approved cancer drugs, while dark pathways are enriched with somatic mutations (or copy number alterations) but not currently targeted by FDA-approved cancer drugs. Our analyses indicated that approximately 35–38% of disease-specific pathways are in scope for repurposing of current cancer drugs. We further assess light and dark pathways for subgroups of patient tumor samples according to HPV status. The framework of light and dark pathways for HNSCC-enriched biological pathways allows us to better prioritize targeted therapies for further research in HNSCC based on the HNSCC genetic landscape and FDA-approved cancer drug information. We also highlight the importance in the identification of sub-pathways where targeting and cross targeting of other pathways may be most beneficial to predict positive or negative synergy with potential clinical significance. This framework is ideal for precision drug panel development, as well as identification of highly aberrant, untargeted candidates for future drug development.
Collapse
Affiliation(s)
- Gabrielle Choonoo
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Aurora S. Blucher
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| | - Samuel Higgins
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Mitzi Boardman
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Sophia Jeng
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Christina Zheng
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - James Jacobs
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
- Pediatric Hematology and Oncology, OHSU Doernbecher Children’s Hospital, Portland, Oregon, United States of America
| | - Ashley Anderson
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Steven Chamberlin
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Nathaniel Evans
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Myles Vigoda
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Benjamin Cordier
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Molly Kulesz-Martin
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Shannon K. McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Ted Laderas
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
35
|
Xu B, Jungbluth AA, Frosina D, Alzumaili B, Aleynick N, Slodkowska E, Higgins K, Ho A, Morris L, Ghossein R, Katabi N. The immune microenvironment and expression of PD-L1, PD-1, PRAME and MHC I in salivary duct carcinoma. Histopathology 2019; 75:672-682. [PMID: 31237963 DOI: 10.1111/his.13944] [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: 05/21/2019] [Accepted: 06/22/2019] [Indexed: 12/11/2022]
Abstract
AIMS Salivary duct carcinoma (SDC) is an aggressive salivary malignancy that results in high mortality rates and is often resistant to chemotherapy. Anti-programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint inhibitors have led to dramatic improvements in patients with various cancers. Other immunotherapeutic approaches, e.g. cancer vaccines, have shown promising results. Cancer testis antigens, e.g. preferentially expressed antigen in melanoma (PRAME), are regarded as promising vaccine targets because of their tumour-specific expression pattern. METHODS AND RESULTS We analysed the immunoexpression of PD-L1, PD-1, major histocompatibility complex class I (MHC I) and PRAME in 53 SDCs. The immunoexpression levels of PD-L1 in tumour cells (TCs) and immune cells (ICs), PD-1 in ICs, PRAME in TCs and MHC I in TCs were analysed, and were correlated with outcome. PRAME expression was seen in 83% of SDCs. No PRAME staining was present in normal salivary gland tissue. With the three established diagnostic algorithms proposed for head and neck squamous cell carcinoma, the criteria being a combined positive score of ≥1, TC% ≥1%, and TC% ≥25%, 35 (66%), 17 (32%) and three cases (6%), respectively, were deemed to be positive for PD-L1. PD-1-positive ICs were seen in 35 (66%) cases. MHC I down-regulation was seen in 82% of SDCs. There was a significant correlation among PD-L1 expression in ICs, PD-1 expression in ICs, and PRAME expression in TCs. PD-L1 expression in TCs and lack of PD-1 expression in ICs were associated with decreased disease-specific survival in SDC patients. CONCLUSIONS Alterations of the tumour immune microenvironment are common in SDCs, including expression of PD-1/PD-L1 and PRAME, which opens the way to potential novel immune therapies, such as cancer vaccination and PD-1/PD-L1 blockade, in these tumours.
Collapse
Affiliation(s)
- Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Denise Frosina
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bayan Alzumaili
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathaniel Aleynick
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elzbieta Slodkowska
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Kevin Higgins
- Department of Otolaryngology-Head & Neck Surgery, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Alan Ho
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
36
|
Huang S, Zhan Z, Li L, Guo H, Yao Y, Feng M, Deng J, Xiong J. LINC00958-MYC positive feedback loop modulates resistance of head and neck squamous cell carcinoma cells to chemo- and radiotherapy in vitro. Onco Targets Ther 2019; 12:5989-6000. [PMID: 31413594 PMCID: PMC6661987 DOI: 10.2147/ott.s208318] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/06/2019] [Indexed: 12/13/2022] Open
Abstract
Background Aberrant long non-coding RNA (lncRNA) expression contributes cancer development and resistance to therapy. This study first assessed expression of lncRNA LINC00958 in a variety of human cancers using GEPIA database data and then associated it with prognosis of head and neck squamous cell carcinoma (HNSCC) and investigated LINC00958 interaction with c-Myc and the c-Myc-related gene interplay in HNSCC cells. Materials and methods A cohort of 48 HNSCC vs normal tissues was collected for qRT-PCR analysis of LINC00958 and c-Myc expression and statistical analyses. HNSCC cell lines were subjected to transfection with LINC00958 and c-Myc siRNAs or cDNA and their negative control siRNA or empty vector for qRT-PCR, Western blot, cell viability, colony formation, luciferase reporter, chromatin immunoprecipitation, and RNA immunoprecipitation assays. Results The data showed that LINC00958 expression was upregulated in HNSCC tissues and cell lines, upregulation of which was associated with poor tumor differentiation, advanced tumor stage, and shorter overall survival of patients. In vitro, LINC00958 expression induced HNSCC cell viability and colony formation, whereas knockdown of LINC00958 expression enhanced HNSCC cell sensitivity to ionizing radiation and cisplatin treatment. Mechanistically, LINC00958 is a direct target of c-Myc and can enhance the transcriptional activity of c-Myc, thus to form a positive feedback gene network in HNSCC cells, and in turn to modulate HNSCC cell resistance to chemo- and radiotherapy. Conclusion This study demonstrated the LINC00958 interplay with c-Myc as a feedback loop facilitated HNSCC development and resistance to chemo- and radiotherapy. Targeting of such a network could be further evaluated as a novel therapeutic strategy for HNSCC patients.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Zhengyu Zhan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Li Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Hui Guo
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Miao Feng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
37
|
PD-L1 Immunohistochemistry Assay Concordance in Urothelial Carcinoma of the Bladder and Hypopharyngeal Squamous Cell Carcinoma. Am J Surg Pathol 2019; 42:1059-1066. [PMID: 29750666 DOI: 10.1097/pas.0000000000001084] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Programmed death ligand-1 (PD-L1) immunohistochemistry is used to guide treatment decisions regarding the use of checkpoint immunotherapy in the management of urothelial carcinoma of the bladder and hypopharyngeal (HP) squamous cell carcinoma. With increasing PD-L1 testing options, a need has arisen to assess the analytical comparability of diagnostic assays in order to develop a more sustainable testing strategy. Using tissue microarrays, PD-L1 expression in tumor cells (TCs) and immune cells (ICs) was manually scored in 197 cases and 27 cases of bladder and HP cancer, respectively. Three commercial kits (Ventana SP263, Ventana SP142, Dako 22C3) and 1 platform-independent test (Cell Signalling Technologies E1L3N) were utilized. Across the 3 commercially available clones, 14% and 74% of urothelial carcinomas were positive and negative, respectively, whereas 7% and 78% of HP carcinomas were positive and negative, respectively. Twelve percent of bladder and 15% HP cases showed discrepant PD-L1 classification results. Regardless of the scoring algorithm used, E1L3N provided comparable PD-L1 staining results. Fleiss' kappa and intraclass correlation coefficient (ICC) analyses demonstrated substantial agreement among all antibody clones (k=0.639 to 0.791) and excellent reliability among SP263, 22C3, and E1L3N antibodies (ICC, 0.929 to 0.949) in TC staining. Compared with the other 3 clones, SP142 TC staining was lower with only moderate correlation (ICC, 0.500 to 0.619). Generally, the reliability of immune cell staining was lower compared with TC staining (ICC, 0.519 to 0.866). Our results demonstrate good analytic comparability of all 4 antibodies. The results are encouraging and support growing optimism in the pathology and oncology communities concerning strategies in PD-L1 assay use.
Collapse
|
38
|
Preusser M. Introducing a new ESMO Open article series: how I treat side effects of immunotherapy. ESMO Open 2019; 4:e000552. [PMID: 31354967 PMCID: PMC6615874 DOI: 10.1136/esmoopen-2019-000552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Affiliation(s)
- Matthias Preusser
- Medical University of Vienna, Department of Medicine I, Division of Oncology, Vienna, Austria.
| |
Collapse
|
39
|
Horton JD, Knochelmann HM, Day TA, Paulos CM, Neskey DM. Immune Evasion by Head and Neck Cancer: Foundations for Combination Therapy. Trends Cancer 2019; 5:208-232. [PMID: 30961829 DOI: 10.1016/j.trecan.2019.02.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/10/2019] [Accepted: 02/15/2019] [Indexed: 12/24/2022]
Abstract
Head and neck cancer is disfiguring and deadly, and contemporary treatment has fallen short in terms of morbidity and mortality. The rich immune infiltrate within these tumors designates them as prime candidates for immunotherapy and success with these drugs has been documented for recurrent and metastatic head and neck cancer. Still, single-agent immunotherapy has generated either only transient responses or durable response in only a minority subset of patients. Mapping the immune escape mechanisms enacted by head and neck cancer within the tumor microenvironment allows for rational design of strategies to overcome this tolerance. We outline the immune pathway derangements within the head and neck cancer microenvironment and discuss combination treatment strategies to overcome the limitations of immunologic monotherapy.
Collapse
Affiliation(s)
- Joshua D Horton
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA.
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA; Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Terry A Day
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA; Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - David M Neskey
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA; Department of Cell and Molecular Pharmacology and Developmental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
40
|
Response to immunotherapy rechallenge after interval chemotherapy in a patient with head and neck cancer. Anticancer Drugs 2019; 30:149-152. [DOI: 10.1097/cad.0000000000000706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Buch S, Chatra L. Immunotherapy and its advances in the management of head-and-neck cancer. CHRISMED JOURNAL OF HEALTH AND RESEARCH 2019. [DOI: 10.4103/cjhr.cjhr_155_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
42
|
Manca P, Raez LE, Salzberg M, Sanchez J, Hunis B, Rolfo C. The value of immunotherapy in head and neck cancer. Expert Opin Biol Ther 2018; 19:35-43. [PMID: 30537444 DOI: 10.1080/14712598.2019.1556637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Head and neck squamous cell carcinomas (HNSCC) previously had limited treatment options once patients had progressed on systemic chemotherapy. With recent advances, immunotherapy now plays an important role in the treatment of advanced disease with improved outcomes as compared to cytotoxic chemotherapy. AREAS COVERED This article reviews the effects of the immune system and how it influences the development and response to HNSCC therapy. We additionally provide a summary of immunotherapy treatments available as well as their applicable clinical trials that led to their approval. EXPERT COMMENTARY The challenges that need to be addressed in order to maximize the benefits of immunotherapy in HNSCC are the selection criteria for immune checkpoint inhibitors and the optimization of combination regimens of immunotherapeutics or chemo-immunotherapy. Furthermore, there remains to be a lack of knowledge in how to incorporate molecular biomarkers as predictors of response to HNSCC immunotherapy.
Collapse
Affiliation(s)
- Paolo Manca
- a Medical Oncology Department , Campus Bio-medico University , Rome , Italy
| | - Luis E Raez
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Matthew Salzberg
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Jorge Sanchez
- c Departamento de Oncologia , Hospital Edgardo Rebagliati Martins , Lima , Peru
| | - Brian Hunis
- b Memorial Cancer Institute/Florida International University , Hollywood , FL , USA
| | - Christian Rolfo
- d Marlene and Stewart Greenebaum Comprehensive Cancer Center , University of Maryland School of Medicine , Baltimore , Maryland , USA
| |
Collapse
|
43
|
Matoba T, Imai M, Ohkura N, Kawakita D, Ijichi K, Toyama T, Morita A, Murakami S, Sakaguchi S, Yamazaki S. Regulatory T cells expressing abundant CTLA-4 on the cell surface with a proliferative gene profile are key features of human head and neck cancer. Int J Cancer 2018; 144:2811-2822. [PMID: 30485427 DOI: 10.1002/ijc.32024] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/07/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
FOXP3+ regulatory T (Treg) cells suppress anti-tumor immunity. The suppression of Treg cells is regulated by cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), whose expression on the cell surface is tightly regulated. Here we found that Treg cells expressing abundant CTLA-4 on the cell surface (surface-CTLA-4+ Treg) were expanded in human head and neck cancer tissues. RNA sequencing of surface-CTLA-4+ and surface-CTLA-4- Treg cells infiltrating human head and neck cancer tissues revealed that surface-CTLA-4+ Treg cells have a previously undescribed gene expression profile correlating to cell cycle, cell proliferation, and DNA replication. Moreover, surface-CTLA-4+ Treg cells were PD-1+ , actively proliferated and associated with CD45RA- FOXP3high Treg cells with strong suppressive function. Thus, surface-CTLA-4+ Treg cells with a proliferative gene expression signature and phenotype are key features of head and neck cancer. Targeting surface-CTLA-4+ Treg cells might be new strategies to evoke effective immune responses to head and neck cancer.
Collapse
Affiliation(s)
- Takuma Matoba
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Oto-rhino-laryngology and Head-and-neck-surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masaki Imai
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Frontier Research in Tumor Immunology, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisuke Kawakita
- Department of Oto-rhino-laryngology and Head-and-neck-surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kei Ijichi
- Department of Oto-rhino-laryngology and Head-and-neck-surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Akaike Oto-rhino-laryngology and Head-and-neck-surgery Clinic, Nisshin, Japan
| | - Tatsuya Toyama
- Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shingo Murakami
- Department of Oto-rhino-laryngology and Head-and-neck-surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Oto-rhino-laryngology, Nagoya City East Medical Center, Nagoya, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Sayuri Yamazaki
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
44
|
Lin W, Chen M, Hong L, Zhao H, Chen Q. Crosstalk Between PD-1/PD-L1 Blockade and Its Combinatorial Therapies in Tumor Immune Microenvironment: A Focus on HNSCC. Front Oncol 2018; 8:532. [PMID: 30519541 PMCID: PMC6258806 DOI: 10.3389/fonc.2018.00532] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/30/2018] [Indexed: 02/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide with a poor prognosis and high mortality. More than two-thirds of HNSCC patients still have no effective control of clinical progression, and the five-year survival rate is < 50%. Moreover, patients with platinum-refractory HNSCC have a median survival of < 6 months. The significant toxicity and low survival rates of current treatment strategies highlight the necessity for new treatment modalities. Recently, a large number of studies have demonstrated that programmed cell death protein-1 (PD-1) and its ligand, programmed cell death protein ligand-1 (PD-L1) play an essential role in tumor initiation and progression. PD-1/PD-L1 blockade has shown a desired and long-lasting therapeutic effect in the treatment of HNSCC and other malignancies. However, only a small number of patients with HNSCC can benefit from PD-1/PD-L1 blockade monotherapy, while the majority of patients do not respond. To overcome the unsatisfactory therapeutic effect of PD-1/PD-L1 blockade monotherapy, combining other treatment options for HNSCC (including chemotherapy, radiotherapy, targeted therapy, and immunotherapy) in the treatment scheme has become a commonly used strategy. Herein, the potential mechanisms underlying the crosstalk between PD-1/PD-L1 blockade and its combinatorial therapies for HNSCC were reviewed, and it is hoped that the improved understanding of the crosstalk process would provide further ideas for the design of a combinatorial regimen with a higher efficiency and response rate for the treatment of HNSCC and other malignancies.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Le Hong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Spielbauer K, Cunningham L, Schmitt N. PD-1 Inhibition Minimally Affects Cisplatin-Induced Toxicities in a Murine Model. Otolaryngol Head Neck Surg 2018; 159:343-346. [PMID: 29609517 PMCID: PMC6134261 DOI: 10.1177/0194599818767621] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/08/2018] [Indexed: 02/01/2023]
Abstract
Immune checkpoint inhibition used in combination with standard cisplatin-based chemotherapy regimens is currently under evaluation in clinical trials for head and neck squamous cell carcinoma (HNSCC). The impact of anti-PD-1 therapy on cisplatin-induced ototoxicity and nephrotoxicity has not been established. Here we use a murine model of cisplatin-induced hearing loss to investigate the impact of anti-PD-1 immunotherapy on auditory brainstem responses (ABRs), distortion product otoacoustic emissions (DPOAEs), serum creatinine, and hair cell and renal histology. We demonstrate only mild worsening of DPOAEs at 14.4 and 16 kHz as well as a mild increase in serum creatinine. Renal and hair cell histology as well as ABR measures were unchanged by PD-1 inhibition. Thus, our data suggest that the use of PD-1 inhibition in conjunction with cisplatin results in toxicities that are similar to those of cisplatin alone.
Collapse
Affiliation(s)
- Katie Spielbauer
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
- Medical Research Scholars Program, National Institutes of Health, Bethesda, Maryland, USA
- Michigan State University College of Human Medicine, East Lansing, Michigan, USA
| | - Lisa Cunningham
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicole Schmitt
- Integrative Therapeutics Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
- Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Denaro N, Merlano MC. Immunotherapy in Head and Neck Squamous Cell Cancer. Clin Exp Otorhinolaryngol 2018; 11:217-223. [PMID: 29973040 PMCID: PMC6222190 DOI: 10.21053/ceo.2018.00150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Prognosis in relapsed metastatic head and neck squamous cell cancer (RM-HNSCC) is dismal. Platinum based chemotherapy in combination with Cetuximab is used in first-line setting, while no further validated options are available at progression. Immunotherapy has produced durable clinical benefit in some patients with RM-HNSCC although the premises are several patients are nonresponders. Studies are ongoing to determine predictive factors and the ideal setting/combination of novel immunotherapies. In this paper, we discuss the past and present of immunotherapy in head and neck cancer and provide an up-to-date information regarding the potential ways to improve immunotherapy outcomes in HNSCC.
Collapse
Affiliation(s)
- Nerina Denaro
- Department of Oncology, ASO Santa Croce e Carle, Cuneo, Italy
| | | |
Collapse
|
47
|
Wolf GT, Moyer JS, Kaplan MJ, Newman JG, Egan JE, Berinstein NL, Whiteside TL. IRX-2 natural cytokine biologic for immunotherapy in patients with head and neck cancers. Onco Targets Ther 2018; 11:3731-3746. [PMID: 29988729 PMCID: PMC6029613 DOI: 10.2147/ott.s165411] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an immunosuppressive malignancy characterized by tumor-driven immune-system abnormalities that contribute to disease progression. For patients with surgically resectable HNSCC, treatment is often curative surgery followed by irradiation or chemoradiation in high-risk settings to reduce the risk of recurrence. Poor survival and considerable morbidity of current treatments suggest the need for new therapeutic modalities that can improve outcomes. Defects in antitumor immunity of HNSCC patients include suppressed dendritic cell (DC) maturation, deficient antigen-presenting cell function, compromised natural killer (NK)-cell cytotoxicity, increased apoptosis of activated T lymphocytes, and impaired immune-cell migration to tumor sites. Strategies for relieving immunosuppression and restoring antitumor immune functions could benefit HNSCC patients. IRX-2 is a primary cell-derived biologic consisting of physiologic levels of T-helper type 1 cytokines produced by stimulating peripheral blood mononuclear cells of normal donors with phytohemagglutinin. The primary active components in IRX-2 are IL2, IL1β, IFNγ, and TNFα. In vitro, IRX-2 acts on multiple immune-system cell types, including DCs, T cells, and NK cells, to overcome tumor-mediated immunosuppression. In clinical settings, IRX-2 is administered as part of a 21-day neoadjuvant regimen, which includes additional pharmacologic agents (low-dose cyclophosphamide, indomethacin, and zinc) to promote anticancer immunoresponses. In a Phase IIA trial in 27 patients with surgically resectable, previously untreated HNSCC, neoadjuvant IRX-2 increased infiltration of T cells, B cells, and DCs into tumors and was associated with radiological reductions in tumor size. Event-free survival was 64% at 2 years, and overall 5-year survival was 65%. Follow-up and data analysis are under way in the multicenter, randomized, Phase IIB INSPIRE trial evaluating the IRX-2 regimen as a stand-alone therapy for activating the immune system to recognize and attack tumors.
Collapse
Affiliation(s)
- Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI,
| | - Jeffrey S Moyer
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI,
| | - Michael J Kaplan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University Medical Center, Stanford, CA
| | - Jason G Newman
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, PA
| | | | | | - Theresa L Whiteside
- Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|