Abstract
In recent years, the field of head and neck oncology has witnessed a remarkable transformation with unprecedented advances that have revolutionized the management of complex tumors in this region. As a critical and challenging subspecialty within oncology, head and neck surgical procedures demand detailed knowledge of the complex anatomy meticulous precision in surgical technique and expertise to preserve vital functions while ensuring optimal oncological outcomes. With the relentless pursuit of improved patient outcomes, the integration of innovative technologies has significantly enhanced the surgical armamentarium. Robotics, endoscopic platforms, and image-guided navigation have revolutionized the surgical approach, enabling precise tumor resection and sparing healthy tissues. Furthermore, the application of advanced imaging modalities and molecular biomarker profiling has opened new avenues for personalized treatment strategies. From targeted therapies and immunotherapies to adaptive radiation techniques, clinicians are now equipped with an array of tailored options, ushering in a new era of personalized care for patients with head and neck malignancies. This article delves into the unfolding narratives of clinical triumphs, exploring the transformative potential of emerging therapies and the collaborative efforts propelling head and neck surgical oncology towards a future of hope and healing.
Introduction
In recent years, the field of head and neck oncology has witnessed a remarkable transformation with unprecedented advances that have revolutionized the management of complex tumors in this region. As a critical and challenging subspecialty within oncology, head and neck surgical procedures demand detailed knowledge of the complex anatomy meticulous precision in surgical technique and expertise to preserve vital functions while ensuring optimal oncological outcomes. With the relentless pursuit of improved patient outcomes, the integration of innovative technologies has significantly enhanced the surgical armamentarium. Robotics, endoscopic platforms, and image-guided navigation have revolutionized the surgical approach, enabling precise tumor resection and sparing healthy tissues. Furthermore, the application of advanced imaging modalities and molecular biomarker profiling has opened new avenues for personalized treatment strategies. From targeted therapies and immunotherapies to adaptive radiation techniques, clinicians are now equipped with an array of tailored options, ushering in a new era of personalized care for patients with head and neck malignancies. This article delves into the unfolding narratives of clinical triumphs, exploring the transformative potential of emerging therapies and the collaborative efforts propelling head and neck surgical oncology towards a future of hope and healing.
Advances in Diagnostic Modalities in Head and Neck Surgery
In the arena of diagnosis of head and neck cancer, we are witnessing an incremental gain to traditional methods of lesion visualization and biopsy with a spectrum of advanced diagnostic techniques. Traditional approaches relied heavily on visual examination and biopsy procedures, but now, innovations like brush biopsy, chemiluminescence, narrow-band imaging (NBI) and auto-fluorescence are ushering in a new era of early detection. Brush biopsy provides a less invasive means to collect cellular samples. An illustrative instance can be found in the research conducted by Goodson et al, where they demonstrated that the utilization of brush biopsy and liquid-based cytology as supplementary methods for handling potentially malignant oral conditions resulted in dependable diagnoses that closely paralleled traditional histopathology assessments. This approach also presented a means to do mass screening in the community and a less invasive means of monitoring patients over the long term.(2) In a parallel fashion, advanced optics using chemiluminescence and auto-fluorescence can accentuate subtle tissue alterations, thereby elevating diagnostic precision, as evidenced in a recent meta-analysis conducted by Moffa et al.(3) Over the past decade, narrow band imaging has become an established adjunct to conventional endoscopic assessment for diagnosing head and neck mucosal cancer.(4,5) In recent years, it has also emerged as a promising tool for identifying a potential primary site of an unknown primary in patients with metastatic neck nodes..(6)
Exploring the frontier of cutting-edge imaging such asPhotoacoustic Imaging (PAI) shines as an exemplary instance. PAI seamlessly integrates ultrasound with laser-induced optical contrast, allowing for the real-time acquisition of high-resolution images. By detecting laser-induced acoustic waves, PAI furnishes essential information about blood vessel distribution and tumor vascularization. A compelling demonstration of this technology’s potential can be observed in the research of Nishio et al., who effectively detected lymph node metastasis in head and neck cancer by synergistically applying PAI with an anti-EGFR (Epidermal Growth Factor Receptor) antibody–dye conjugate.(7) In this context, EGFR serves as a prime example of molecular-based imaging in head and neck surgery. Molecular imaging techniques have also been used to monitor efficacy of head and neck cancer treatment. Antibodies like cetuximab and panitumumab have been labeled with various radionuclides for non-invasive imaging, allowing for therapy monitoring and personalized treatment planning.(8,9) These imaging methods have provided insights into EGFR expression and drug accessibility in tumors. Combining diagnostic and therapeutic potential, theranostic strategies are being explored, showing promise in target selection and therapy.
Nanotechnology has also played an important role in enhancing diagnostic accuracy. Engineered nanoparticles can target specific cancer markers, allowing for early detection through imaging methods like MRI and PET. These advanced materials, including phospholipid nanomicelles, gold nanoparticles, graphene, quantum dots, and hydrogels, enhance head and neck imaging by providing better contrast and enabling optical image-guided surgery.(10) In addition, nanosensors, similar in scale to biological systems, are being developed, targeting specific biomarkers and genes associated with head and neck cancer. Researchers have created effective nanosensors, like Au nanorattles(11) and electro-chemiluminescence detectors,(12) to spot specific markers with remarkable precision. These nanosensors also show promise for detecting squamous cell carcinoma antigen(13) and HPV markers,(14) potentially improving early cancer diagnosis.
As a final noteworthy development, transcending traditional imaging methods, metabolomics, which examines metabolic changes in the body, has introduced techniques like Magnetic Resonance Spectroscopy(15) and Hyperpolarized MRI.(16) These methods offer insights into biochemical alterations related to head and neck cancers. By detecting shifts in metabolite profiles, they help differentiate between benign and malignant lesions, predict treatment responses, and support personalized treatment plans.
Technological advances in the Surgical Arena
The landscape of head and neck surgery is undergoing a dynamic transformation fueled by cutting-edge technologies and evidence-based innovations. Recent advancements are orchestrating a journey that begins with enhanced visualization techniques and navigational precision, leading to patient-centered procedures, all while harnessing the power of data-driven insights.
The conventional use of endoscopes and microscopes has paved the way for a revolutionary leap in visualization. Exoscopy has emerged as a transformative tool employin high-definition cameras and advanced optics offering magnified visualization of the surgical field in minimally invasive procedures.(17) By utilizing exoscopic systems, surgeons can access anatomically challenging areas with greater ease and accuracy, reducing the need for extensive tissue dissection. Furthermore, exoscopy enhances the educational aspect of surgery by enabling real-time sharing of surgical field with colleagues and trainees.(18) With its potential to optimize surgical outcomes, minimize patient discomfort, and facilitate skill transfer, exoscopy is poised to play an increasing role in advancing the field of head and neck surgery. Within this field, in addition to enhanced visualization, technologies like Augmented Reality (AR) have ushered in a new era of precision. AR’s potential is particularly evident in skull base surgery. This approach provides surgeons with an augmented, three-dimensional view of the surgical field.(19) By superimposing critical anatomical structures, such as, blood vessels, orbits, and brain, onto the surgeon’s real-time visual field, AR offers unparalleled precision and spatial awareness. In their systematic review, Thavarajasingam et al. found that augmented reality improves landmark identification, intraoperative navigation, and surgeon experience in transsphenoidal surgery, enhancing accuracy and efficiency.(20) Moreover, AR technology enriches the educational opportunities for students and trainees, providing immersive, practical learning experiences, as demonstrated in a recent study by Weeks and colleagues.(21)
Intraoperative imaging methods continue to improve the detection of tumor margins, thereby facilitating more precise tumor removal. A systematic review conducted by Ravin et al. highlighted the efficacy of AB-dye conjugate imaging in achieving remarkable specificity and accuracyin tumor localization and resection.(22) This innovative technique, in conjunction with fluorescence-based imaging utilizing indocyanine green, facilitates precise delineation of tumors, potentially minimizing the requirement for extensive resections and thereby improving patient outcomes. In line with this, De Wit and colleagues recently published the results of a phase II trial focusing on EGFR-targeted fluorescence molecular imaging for intraoperative margin assessment in oral cancer patients. Their findings revealed 100% sensitivity in detecting tumor-positive margins along with a specificity of 85.9%, potentially paving the way for more precise tumor resection with reduced incidence of positive margins.(23)
Last but certainly not least, is the integration of robotics in head and neck surgeryover the past decade, has seen significant. Transoral Robotic Surgery (TORS) has become a prominent choice in minimally invasive procedures, particularly for early-stage oropharyngeal tumors.(24,25) The introduction of flexible endoscope systems like the Flex System has improved accessibility and cost-effectiveness,(26) while the latest DaVinci Single-Port system holds promise for enhanced visualization and refined instrument handling.(27) In skull base and paranasal sinus surgery, challenges remain, but there’s promise in developing flexible robotic systems.(28) Within the realm of thyroid and neck surgeries, where aesthetics play a significant role, the introduction of robot-assisted procedures, such as transoral neck dissection and thyroidectomy, have generated interest.(29,30) Nevertheless, even though small retrospective studies have indicated that robot-assisted surgery yields outcomes comparable to those of traditional open surgery,(31,32) a critical gap exists in terms of comprehensive data and large-scale randomized trials assessing overall survival and disease-free survival, as well as cost effectiveness of these procedure, potentialcomplications and sequela and eventually patient satisfaction.. Case selection for robotic surgery is an important factor, which impacts on outcome. This circumstance further accentuates the intricate ethical dilemma concerning the delicate equilibrium between aesthetics and oncologic safety. TORS on the other hand, presents compelling advantages in the excision of retropharyngeal lymph nodes, as it furnishes histological confirmation of metastases. This capability holds the potential to prevent the administration of unnecessary adjuvant treatments.(33,34)
Evolving Strategies Beyond the Scalpel
While surgical intervention remains a cornerstone in the management of head and neck malignancies, an evolving understanding of the disease biology and a desire to minimize treatment-related morbidity have prompted the exploration of non-surgical approaches.
The management of oropharyngeal cancer, frequently associated with human papillomavirus (HPV) infection, has witnessed a paradigm shift towards de-escalation of treatment intensity. Traditionally, patients with oropharyngeal cancer underwent extensive surgery and radiation therapy, leading to substantial morbidity in terms of swallowing and speech function. However, accumulating empirical evidence suggests that not all patients necessitate such aggressive therapeutic approaches. In pursuit of a patient-centered care model, de-escalation strategies have assumed a central role. These approaches prioritize non-surgical interventions and aim to tailor treatment based on individual patient and tumor risk profiles. Such customization may involve reducing radiation dosages or exploring less invasive procedures. Noteworthy examples include the AVOID trial, which specifically examined neck-only adjuvant radiotherapy for select patients, yielding impressive 2-year progression-free and overall survival rates.(35) Trials such as ECOG-ACRIN 3311 and PATHOS have similarly hinted at potential benefits associated with reduced-dose adjuvant radiation therapy for intermediate-risk patients in comparison to standard protocols.(36,37) While techniques like TORS have proven valuable in this endeavor, the broader emphasis lies in the pursuit of tailored non-surgical solutions that minimize the impact on patients’ quality of life. Several clinical trials have explored reduced-intensity chemoradiation, contingent upon responses to induction chemotherapy, yielding variable outcomes. Ongoing trials such as OPTIMA-II and Quarterback IIb continue to investigate response-based de-escalation following induction chemotherapy.(38,39) The integration of advanced imaging modalities and molecular profiling has further empowered clinicians to identify low-risk patients who can derive benefit from these less aggressive treatment modalities. An illustrative example of this approach can be found in the ongoing 30 ROC trial by Lee et al. from MSKCC. Their recent update, which involved significant de-escalation to a 30Gy radiation dose based on patient-specific treatment response criteria, particularly hypoxia resolution, has yielded excellent locoregional control while significantly reducing treatment-related toxicity.(40)
The concept of de-escalation has been gradually gaining traction in various aspects of head and neck surgical oncology, with thyroid cancer standing out as a notable example. Over the past decade, the approach of active surveillance has garnered significant attention in managing differentiated thyroid cancer, especially among patients with low-risk tumors. A recent systematic review conducted by Chou et al. has suggested that for individuals with small, low-risk differentiated thyroid cancer, active surveillance and immediate surgery may have similar outcomes in terms of mortality and recurrence risk.(41) This approach has garnered significant global backing, as illustrated, for instance, by the consensus declarations issued by the Japanese thyroid and endocrine surgery associations in their 2021 publications.(42,43) In recent years, monitoring low-risk thyroid cancer patients has seen continuous advancements. Notably, a study led by Tuttle and Morris at MSKCC introduced a six-pattern tumor volume kinetic classification system, which assists in discerning actionable findings from mere observations and thereby supports personalized management decisions.(44)
The other extreme of non-surgical treatments encompasses cancers that are recurrent, metastatic, or inoperable. In such circumstances, immunotherapy is steadily earning recognition as a preferred choice for treatment. Pembrolizumab, a PD-1 monoclonal antibody, made groundbreaking strides as the first immunotherapy for recurrent or metastatic head and neck squamous cell carcinoma.(45) The KEYNOTE studies, spanning phases I to III, meticulously assessed its efficacy and safety.(46–48) Harrington et al. recently reported updated results from the phase III KEYNOTE-048 study, showing that even after a 4-year follow-up, first-line pembrolizumab and pembrolizumab-chemotherapy still provided better survival outcomes compared to cetuximab-chemotherapy in recurrent/metastatic head and neck squamous cell carcinoma.(49) In addition to pembrolizumab, nivolumab, another PD-1 antibody, received FDA approval for recurrent and metastatic head and neck cancer based on the results of the CheckMate 141 trial.(50) These developments, supported by ongoing research, not only enhanced overall and progression-free survival but also opened doors for agents like durvalumab, atezolizumab, and avelumab to combat far advanced head and neck cancer.(51–53) This progress underscores PD-1/PD-L1 blockade’s potential in restoring anti-tumor immune responses, marking a pivotal shift in head and neck cancer care.
Beyond PD-1/PD-L1 blockade, several other immunotherapeutic strategies have been explored. These include targeting regulatory T cells (Tregs), anti-TIM-3 and anti-LAG-3 agents, monoclonal antibodies blocking NKG2A, and therapies focused on inducible T cell co-stimulator and inducible T cell co-stimulator ligand (ongoing INDUCE-3 and INDUCE-4 trials).(54–57) These approaches aim to enhance the anti-tumor immune response and broaden the scope of treatment options. Transitioning from the context of more established immunotherapy treatments, a shift occurs towards the forefront of targeted therapies. Three noteworthy contenders in this arena are tumor vaccines, adoptive cellular immunotherapy, and oncolytic viruses. Prophylactic vaccines have demonstrated success in preventing precursor lesions, particularly in HPV-associated head and neck cancers.(58) Meanwhile, therapeutic vaccines, when combined with immune checkpoint inhibitors, have shown potential in boosting T cell responses.(59) Adoptive cellular immunotherapy, including chimeric antigen receptor T cell (CAR-T cell) therapy, offers a personalized approach to cancer treatment. While showing promise, CAR-T cell therapy is currently limited by significant toxicity and remains an evolving technique.(60) Oncolytic viruses, such as T-VEC, present a novel avenue for selectively targeting and destroying tumor cells. Although their potential in head and neck cancer treatment is still being explored, combining oncolytic viruses with other immunotherapeutic agents holds promise.(59)
Beyond Treatment: Innovations in Monitoring Head and Neck Cancer
The realm of head and neck surgical oncology extends well beyond the treatment phase. This section focuses on recent advancems in post-treatment surveillance, highlighting innovations in how patients are monitored during the follow up period. From predictive markers such as tumor mutational burden (TMB), and circulating tumorDNA (ctDNA) as in nasopharynx and HPV associated oropharynx cancers, to advanced imaging techniques, these developments objectively redefine and optimize the approach to post-treatment follow-up in this intricate field.
One of the most exciting developments in head and neck oncology is the use of biomarkers such as Tumoral Mutational Burden (TMB) to predict treatment response. TMB quantifies the number of mutations within a tumor, providing valuable insights into its genomic instability. High TMB has been associated with increased responsiveness to immunotherapy, particularly immune checkpoint inhibitors.(61,62) However, as data continue to accrue, it becomes evident that considerations extend beyond the concept of TMB alone. Factors such as immune cell infiltration, the pre-treatment neutrophil-to-lymphocyte ratio, and a spectrum of other variables are now recognized for their roles in determining the response to therapy.(63,64)
The quest for precise biomarkers has propelled the rise of Next-Generation Sequencing (NGS), a sophisticated technology for cost-effective DNA and RNA sequencing.(65) NGS has ushered in a new era in head and neck cancer research, uncovering novel genetic mutations and molecular signatures while enabling comprehensive analysis of tumor genomics. NGS platforms range from dissecting single-gene anomalies, such as EGFR and BRAF mutations, to comprehensive panels like FoundationOne CDx.(66,67) In the context of head and neck cancer, the preference for multi-gene sequencing over isolated gene testing is evident. This preference not only enables the exploration of multiple actionable targets but also eliminates the need for repetitive biopsies, thus averting potential treatment delays.(68) The primary objective is to efficiently direct patients towards the most suitable clinical trials when applicable. Furthermore, some clinicians have started incorporating NGS into routine care to establish correlations between genomic alterations and oncological outcomes.(69)
NGS can also be applied to detect numerous fragments of circulating tumor DNA (ctDNA) present in blood and saliva samples, commonly referred to as “liquid biopsies.”(70,71) In this context, the distinctive molecular imprint of HPV associated oropharyngeal cancer rendering it an exceptionally promising contender for the monitoring and detection of HPV-mediated disease. Thanks to the technological progress, in the sensitive and dependable quantification and detection of HPV gene expression at minimal copy numbers in the bloodstream have become possible, as reflected in the commercially accessible NavDx assessment (Naveris).(72) In a recently published clinical investigation carried out by Ferrandino et al., the researchers employed the NavDx assay, revealing a specificity of 100% and a sensitivity of 88.4% in the surveillance of HPV-associated oropharyngeal carcinoma.(73) The NavDx assay’s promise in monitoring HPV-related oropharyngeal carcinoma highlights how ctDNA can revolutionize cancer diagnosis and monitoring, pointing toward a future where liquid biopsies enhance the precision and sensitivity of cancer detection and management,
Finally, the field of monitoring head and neck cancer patients appears to be on the cusp of a new era, characterized by the seamless integration of computer technology, big data and artificial intelligence (AI) to enhance patient care. Initiatives like BD4QoL, a multicenter randomized trial employing non-invasive intelligent tools for quality-of-life assessment, underscore the commitment of healthcare professionals to address not only the physical aspects of patient well-being but also to explore the intricate dimensions of their overall quality of life.(74) AI, in the form of deep learning and convolutional neural networks, is also harnessed for early tumor detection, classification, prognosis, and metastasis prediction.(75). As technology and research advancements progress, the future promises significant potential for enhancing outcomes and elevating the quality of life for individuals battling head and neck cancer.
Forging a Path to Enhanced Communication and Patient Care in Head and Neck Surgical Oncology
In the field of head and neck oncology, advances in technology and patient-centered care have been instrumental in improving, outcomes and enhancing the overall quality of healthcare delivery. The COVID-19 pandemic, although challenging, has spurred innovation in healthcare, particularly in the area of communication with patients and telemedicine. This section explores the remarkable progress made in communication strategies, the integration of telecommunication, the refinement of video chats and examinations, and the implementation of patient feedback systems, all of which have led to enhanced patient experiences and improved, efficient and cost effective healthcare provider performance.
The onset of the COVID-19 pandemic propelled the swift integration of telecommunication and telemedicine into the practice of medicine in general and head and neck onology in particular. Telemedicine platforms have empowered healthcare providers to conduct virtual consultations, thereby diminishing the necessity for in-person visits. This shift enables patients to conveniently discuss their concerns, receive updates, and obtain medical advice from the comfort of their homes.(76) Furthermore, the incorporation of high-definition video technology has enabled meticulous visual examinations, affording healthcare providers the ability to assess surgical sites and monitor postoperative healing remotely. This technological innovation has yielded enhancements in diagnostic precision and contributed to a reduction in the requirement for physical examinations, as evidenced in a study that compared video-based free flap assessment following head and neck free tissue transfer to in-person evaluations.(77)
Over the past ten years, shared decision-making models have risen in prominence, enabling patients to take an active role in shaping their treatment plans. Physicians now furnish patients with comprehensive details regarding the array of treatment options, potential risks, and benefits, thereby guaranteeing that patients have a say in the formulation of their care strategies.(78) Simultaneously, the advent of remote monitoring devices and applications has granted patients the capacity to actively engage in their healthcare journey by closely monitoring vital signs, tracking side effects, and observing the progress of their recovery. This technology equips healthcare providers with the means to promptly intervene in the event of issues arising, thereby fostering early intervention and ultimately yielding improved outcomes.(79)
Finally, in the past decade the field of head and neck oncology has witnessed significant progress, prompting healthcare professionals to steadfastly embrace the ongoing integration of cultural competency training. This unwavering commitment ensures that patient care remains adaptable, finely tuned to meet the distinct needs, beliefs, and preferences of each individual, thus continually enhancing the relationship between patients and providers. An illustrative example of this sustained effort is seen in initiatives such as the Royal Australasian College of Surgeons’ development of a Māori Health Action Plan,(80) as well as in notable publications consistently emphasizing the critical importance of diversity within otolaryngology–head and neck surgery, particularly in relation to underrepresented minority communities.(81)
Final Insights: Charting Head and Neck Oncology’s Promising Path
In the world of head and neck surgical oncology, the past decade has witnessed a profound fusion of technology and patient-centered care. Advancements in diagnostics, surgical techniques, and treatment options have collectively ushered in a new era of precision medicine. Looking ahead to the next decade, we anticipate a continued evolution characterized by even more personalized approaches, minimally invasive surgical innovations, and an expanding range of targeted therapies. These developments hold the promise of a future where head and neck cancer management is characterized by greater individualization, reduced invasiveness, and improved patient experiences. Moreover, the lessons learned from the COVID-19 pandemic have accelerated the integration of telemedicine and shared decision-making, poised to become integral components of care in the years to come. As we navigate this ever-changing landscape, our unwavering commitment to excellence and patient well-being remains our guiding force, ensuring that each patient’s journey is marked by progress and compassion.
Synopsis:
A major advance in the management of Head and Neck tumors is the introduction of technology such as NBI and autofluorescence in diagnosis and robotics, navigation and minimally invasive techniques in resection as well as CAD-CAM in reconstruction. Treatment deintensification to reduce morbidity is the focus in HPV positive cancers, and the role of immunotherapy is under intense investigation to improve long term outcomes.
References
- 1.Chhabra N, Chhabra S, Sapra N. Diagnostic modalities for squamous cell carcinoma: an extensive review of literature-considering toluidine blue as a useful adjunct. J Maxillofac Oral Surg. 2015. Jun;14(2):188–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Goodson ML, Smith DR, Thomson PJ. Efficacy of oral brush biopsy in potentially malignant disorder management. J Oral Pathol Med. 2017. Nov;46(10):896–901. [DOI] [PubMed] [Google Scholar]
- 3.Moffa A, Giorgi L, Costantino A, De Benedetto L, Cassano M, Spriano G, et al. Accuracy of autofluorescence and chemiluminescence in the diagnosis of oral Dysplasia and Carcinoma: A systematic review and Meta-analysis. Oral Oncol. 2021. Oct;121:105482. [DOI] [PubMed] [Google Scholar]
- 4.Muto M, Minashi K, Yano T, Saito Y, Oda I, Nonaka S, et al. Early Detection of Superficial Squamous Cell Carcinoma in the Head and Neck Region and Esophagus by Narrow Band Imaging: A Multicenter Randomized Controlled Trial. J Clin Oncol. 2010. Mar 20;28(9):1566–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhou H, Zhang J, Guo L, Nie J, Zhu C, Ma X. The value of narrow band imaging in diagnosis of head and neck cancer: a meta-analysis. Sci Rep. 2018. Jan 11;8:515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Di Maio P, Iocca O, De Virgilio A, Giudice M, Pellini R, D’Ascanio L, et al. Narrow band imaging in head and neck unknown primary carcinoma: A systematic review and meta-analysis. Laryngoscope. 2020. Jul;130(7):1692–700. [DOI] [PubMed] [Google Scholar]
- 7.Nishio N, van den Berg NS, Martin BA, van Keulen S, Fakurnejad S, Rosenthal EL, et al. Photoacoustic Molecular Imaging for the Identification of Lymph Node Metastasis in Head and Neck Cancer Using an Anti-EGFR Antibody-Dye Conjugate. J Nucl Med. 2021. May 10;62(5):648–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Even AJG, Hamming-Vrieze O, van Elmpt W, Winnepenninckx VJL, Heukelom J, Tesselaar MET, et al. Quantitative assessment of Zirconium-89 labeled cetuximab using PET/CT imaging in patients with advanced head and neck cancer: a theragnostic approach. Oncotarget. 2017. Jan 17;8(3):3870–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wu J, Yuan Y, Tao XF. Targeted molecular imaging of head and neck squamous cell carcinoma: a window into precision medicine. Chin Med J (Engl). 2020. Jun 5;133(11):1325–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ruiz-Pulido G, Medina DI, Barani M, Rahdar A, Sargazi G, Baino F, et al. Nanomaterials for the Diagnosis and Treatment of Head and Neck Cancers: A Review. Materials (Basel). 2021. Jul 2;14(13):3706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Vohra P, Strobbia P, Ngo HT, Lee WT, Vo-Dinh T. Rapid Nanophotonics Assay for Head and Neck Cancer Diagnosis. Sci Rep. 2018. Jul 30;8(1):11410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wang Y, Jiang M, Shan Y, Jin X, Gong M, Wang X. Nano polythionine-based electrochemiluminescence biosensor for detection of the p16INK4a gene using RuAg@AuNPs core-shell nanocomposites as DNA labels. Journal of Luminescence. 2018. Sep 1;201:135–42. [Google Scholar]
- 13.Wang Y, Guo Y, Lu J, Sun Y, Yu X, Gopinath SCB, et al. Nanodetection of Head and Neck Cancer on Titanium Oxide Sensing Surface. Nanoscale Res Lett. 2020. Feb 3;15(1):33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yokchom R, Laiwejpithaya S, Maneeprakorn W, Tapaneeyakorn S, Rabablert J, Dharakul T. Paper-based immunosensor with signal amplification by enzyme-labeled anti-p16INK4a multifunctionalized gold nanoparticles for cervical cancer screening. Nanomedicine. 2018. Apr;14(3):1051–8. [DOI] [PubMed] [Google Scholar]
- 15.Fujima N, Carlota Andreu-Arasa V, Barest GD, Srinivasan A, Sakai O. Magnetic Resonance Spectroscopy of the Head and Neck: Principles, Applications, and Challenges. Neuroimaging Clin N Am. 2020. Aug;30(3):283–93. [DOI] [PubMed] [Google Scholar]
- 16.Mignion L, Acciardo S, Gourgue F, Joudiou N, Caignet X, Goebbels RM, et al. Metabolic Imaging Using Hyperpolarized Pyruvate-Lactate Exchange Assesses Response or Resistance to the EGFR Inhibitor Cetuximab in Patient-Derived HNSCC Xenografts. Clin Cancer Res. 2020. Apr 15;26(8):1932–43. [DOI] [PubMed] [Google Scholar]
- 17.Paderno A, Deganello A, Lancini D, Piazza C. Is the exoscope ready to replace the operative microscope in transoral surgery? Curr Opin Otolaryngol Head Neck Surg. 2022. Apr 1;30(2):79–86. [DOI] [PubMed] [Google Scholar]
- 18.De Virgilio A, Iocca O, Di Maio P, Mercante G, Mondello T, Yiu P, et al. Free flap microvascular anastomosis in head and neck reconstruction using a 4K three-dimensional exoscope system (VITOM 3D). Int J Oral Maxillofac Surg. 2020. Sep;49(9):1169–73. [DOI] [PubMed] [Google Scholar]
- 19.Rose AS, Kim H, Fuchs H, Frahm JM. Development of augmented-reality applications in otolaryngology-head and neck surgery. Laryngoscope. 2019. Oct;129 Suppl 3:S1–11. [DOI] [PubMed] [Google Scholar]
- 20.Thavarajasingam SG, Vardanyan R, Arjomandi Rad A, Thavarajasingam A, Khachikyan A, Mendoza N, et al. The use of augmented reality in transsphenoidal surgery: A systematic review. Br J Neurosurg. 2022. Aug;36(4):457–71. [DOI] [PubMed] [Google Scholar]
- 21.Weeks JK, Pakpoor J, Park BJ, Robinson NJ, Rubinstein NA, Prouty SM, et al. Harnessing Augmented Reality and CT to Teach First-Year Medical Students Head and Neck Anatomy. Acad Radiol. 2021. Jun;28(6):871–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.De Ravin E, Venkatesh S, Harmsen S, Delikatny EJ, Husson MA, Lee JYK, et al. Indocyanine green fluorescence-guided surgery in head and neck cancer: A systematic review. Am J Otolaryngol. 2022;43(5):103570. [DOI] [PubMed] [Google Scholar]
- 23.de Wit JG, Vonk J, Voskuil FJ, de Visscher SAHJ, Schepman KP, Hooghiemstra WTR, et al. EGFR-targeted fluorescence molecular imaging for intraoperative margin assessment in oral cancer patients: a phase II trial. Nat Commun. 2023. Aug 16;14(1):4952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Park YM, Jung CM, Cha D, Kim SH. The long-term oncological and functional outcomes of transoral robotic surgery in patients with hypopharyngeal cancer. Oral Oncol. 2017. Aug;71:138–43. [DOI] [PubMed] [Google Scholar]
- 25.Hussain T, Lang S, Haßkamp P, Holtmann L, Höing B, Mattheis S. The Flex robotic system compared to transoral laser microsurgery for the resection of supraglottic carcinomas: first results and preliminary oncologic outcomes. Eur Arch Otorhinolaryngol. 2020. Mar;277(3):917–24. [DOI] [PubMed] [Google Scholar]
- 26.Sethi N, Gouzos M, Padhye V, Ooi EH, Foreman A, Krishnan S, et al. Transoral robotic surgery using the Medrobotic Flex® system: the Adelaide experience. J Robot Surg. 2020. Feb;14(1):109–13. [DOI] [PubMed] [Google Scholar]
- 27.Holsinger FC, Magnuson JS, Weinstein GS, Chan JYK, Starmer HM, Tsang RKY, et al. A Next-Generation Single-Port Robotic Surgical System for Transoral Robotic Surgery: Results From Prospective Nonrandomized Clinical Trials. JAMA Otolaryngol Head Neck Surg. 2019. Nov 1;145(11):1027–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wang EW, Zanation AM, Gardner PA, Schwartz TH, Eloy JA, Adappa ND, et al. ICAR: endoscopic skull-base surgery. Int Forum Allergy Rhinol. 2019. Jul;9(S3):S145–365. [DOI] [PubMed] [Google Scholar]
- 29.Tae K, Kim KH. Transoral robotic selective neck dissection for papillary thyroid carcinoma: Dissection of Levels III and IV. Head Neck. 2020. Oct;42(10):3084–8. [DOI] [PubMed] [Google Scholar]
- 30.Lira RB, Ramos AT, Nogueira RMR, de Carvalho GB, Russell JO, Tufano RP, et al. Transoral thyroidectomy (TOETVA): Complications, surgical time and learning curve. Oral Oncol. 2020. Nov;110:104871. [DOI] [PubMed] [Google Scholar]
- 31.You JY, Kim HY, Chai YJ, Kim HK, Anuwong A, Tufano RP, et al. Transoral Robotic Thyroidectomy Versus Conventional Open Thyroidectomy: Comparative Analysis of Surgical Outcomes in Thyroid Malignancies. J Laparoendosc Adv Surg Tech A. 2019. Jun;29(6):796–800. [DOI] [PubMed] [Google Scholar]
- 32.Sukato DC, Ballard DP, Abramowitz JM, Rosenfeld RM, Mlot S. Robotic versus conventional neck dissection: A systematic review and meta-analysis. Laryngoscope. 2019. Jul;129(7):1587–96. [DOI] [PubMed] [Google Scholar]
- 33.Ding X, Lin QG, Zou X, Liu YP, Hua YJ, Xie YL, et al. Transoral Robotic Retropharyngeal Lymph Node Dissection in Nasopharyngeal Carcinoma With Retropharyngeal Lymph Node Recurrence. Laryngoscope. 2021. Jun;131(6):E1895–902. [DOI] [PubMed] [Google Scholar]
- 34.Garas G, Roland NJ, Lancaster J, Zammit M, Manon VA, Davies K, et al. Novel Strategies for Managing Retropharyngeal Lymph Node Metastases in Head and Neck and Thyroid Cancer with Transoral Robotic Surgery (TORS). Ann Surg Oncol. 2022. Nov;29(12):7881–90. [DOI] [PubMed] [Google Scholar]
- 35.Swisher-McClure S, Lukens JN, Aggarwal C, Ahn P, Basu D, Bauml JM, et al. A Phase 2 Trial of Alternative Volumes of Oropharyngeal Irradiation for De-intensification (AVOID): Omission of the Resected Primary Tumor Bed After Transoral Robotic Surgery for Human Papilloma Virus-Related Squamous Cell Carcinoma of the Oropharynx. Int J Radiat Oncol Biol Phys. 2020. Mar 15;106(4):725–32. [DOI] [PubMed] [Google Scholar]
- 36.Ferris RL, Flamand Y, Weinstein GS, Li S, Quon H, Mehra R, et al. Phase II Randomized Trial of Transoral Surgery and Low-Dose Intensity Modulated Radiation Therapy in Resectable p16+ Locally Advanced Oropharynx Cancer: An ECOG-ACRIN Cancer Research Group Trial (E3311). J Clin Oncol. 2022. Jan 10;40(2):138–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Owadally W, Hurt C, Timmins H, Parsons E, Townsend S, Patterson J, et al. PATHOS: a phase II/III trial of risk-stratified, reduced intensity adjuvant treatment in patients undergoing transoral surgery for Human papillomavirus (HPV) positive oropharyngeal cancer. BMC Cancer. 2015. Aug 27;15:602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Seiwert TY, Foster CC, Blair EA, Karrison TG, Agrawal N, Melotek JM, et al. OPTIMA: a phase II dose and volume de-escalation trial for human papillomavirus-positive oropharyngeal cancer. Ann Oncol. 2019. Feb 1;30(2):297–302. [DOI] [PubMed] [Google Scholar]
- 39.Posner MR, Misiukiewicz K, Miles BA, Sharma S, Gupta V, Genden EM, et al. Survival (OS) and progression-free survival (PFS) results after induction chemotherapy (IC) followed by de-escalated chemoradiotherapy (RDCRT) for locally advanced (LA) HPV positive oropharynx cancer (HPVOPC. JCO. 2021. May 20;39(15_suppl):6058–6058. [Google Scholar]
- 40.Lee NY, Sherman EJ, Schöder H, McBride SM, Yu Y, Kang J, et al. The 30 ROC trial: Precision intra-treatment imaging guiding major radiation reduction in human papillomavirus related oropharyngeal cancer. JCO. 2021. May 20;39(15_suppl):6019–6019. [Google Scholar]
- 41.Chou R, Dana T, Haymart M, Leung AM, Tufano RP, Sosa JA, et al. Active Surveillance Versus Thyroid Surgery for Differentiated Thyroid Cancer: A Systematic Review. Thyroid. 2022. Apr;32(4):351–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Horiguchi K, Yoshida Y, Iwaku K, Emoto N, Kasahara T, Sato J, et al. Position paper from the Japan Thyroid Association task force on the management of low-risk papillary thyroid microcarcinoma (T1aN0M0) in adults. Endocr J. 2021. Jul 28;68(7):763–80. [DOI] [PubMed] [Google Scholar]
- 43.Sugitani I, Ito Y, Takeuchi D, Nakayama H, Masaki C, Shindo H, et al. Indications and Strategy for Active Surveillance of Adult Low-Risk Papillary Thyroid Microcarcinoma: Consensus Statements from the Japan Association of Endocrine Surgery Task Force on Management for Papillary Thyroid Microcarcinoma. Thyroid. 2021. Feb;31(2):183–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tuttle RM, Fagin J, Minkowitz G, Wong R, Roman B, Patel S, et al. Active Surveillance of Papillary Thyroid Cancer: Frequency and Time Course of the Six Most Common Tumor Volume Kinetic Patterns. Thyroid. 2022. Nov;32(11):1337–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pereira D, Martins D, Mendes F. Immunotherapy in Head and Neck Cancer When, How, and Why? Biomedicines. 2022. Sep 1;10(9):2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, et al. Pembrolizumab for Platinum- and Cetuximab-Refractory Head and Neck Cancer: Results From a Single-Arm, Phase II Study. J Clin Oncol. 2017. May 10;35(14):1542–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mehra R, Seiwert TY, Gupta S, Weiss J, Gluck I, Eder JP, et al. Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012. Br J Cancer. 2018. Jul;119(2):153–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G, et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet. 2019. Nov 23;394(10212):1915–28. [DOI] [PubMed] [Google Scholar]
- 49.Harrington KJ, Burtness B, Greil R, Soulières D, Tahara M, de Castro G, et al. Pembrolizumab With or Without Chemotherapy in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Updated Results of the Phase III KEYNOTE-048 Study. J Clin Oncol. 2023. Feb 1;41(4):790–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ferris RL, Blumenschein G, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab vs investigator’s choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral Oncol. 2018. Jun;81:45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ferris RL, Haddad R, Even C, Tahara M, Dvorkin M, Ciuleanu TE, et al. Durvalumab with or without tremelimumab in patients with recurrent or metastatic head and neck squamous cell carcinoma: EAGLE, a randomized, open-label phase III study. Ann Oncol. 2020. Jul;31(7):942–50. [DOI] [PubMed] [Google Scholar]
- 52.Bhatia AK, Flamand Y, Johnson JM, Ishizuka JJ, Duan F, Tang M, et al. A phase II/III trial of chemotherapy plus cetuximab versus chemotherapy plus bevacizumab versus atezolizumab plus bevacizumab following progression on immune checkpoint inhibition in recurrent/metastatic head and neck cancers: ECOG-ACRIN EA3202. JCO. 2022. Jun;40(16_suppl):TPS6098–TPS6098. [Google Scholar]
- 53.Lee NY, Ferris RL, Psyrri A, Haddad RI, Tahara M, Bourhis J, et al. Avelumab plus standard-of-care chemoradiotherapy versus chemoradiotherapy alone in patients with locally advanced squamous cell carcinoma of the head and neck: a randomised, double-blind, placebo-controlled, multicentre, phase 3 trial. Lancet Oncol. 2021. Apr;22(4):450–62. [DOI] [PubMed] [Google Scholar]
- 54.Castillo J, Wu E, Lowe C, Srinivasan S, McCord R, Wagle MC, et al. CBP/p300 Drives the Differentiation of Regulatory T Cells through Transcriptional and Non-Transcriptional Mechanisms. Cancer Res. 2019. Aug 1;79(15):3916–27. [DOI] [PubMed] [Google Scholar]
- 55.Fasano M, Corte CMD, Liello RD, Viscardi G, Sparano F, Iacovino ML, et al. Immunotherapy for head and neck cancer: Present and future. Crit Rev Oncol Hematol. 2022. Jun;174:103679. [DOI] [PubMed] [Google Scholar]
- 56.van Hall T, André P, Horowitz A, Ruan DF, Borst L, Zerbib R, et al. Monalizumab: inhibiting the novel immune checkpoint NKG2A. J Immunother Cancer. 2019. Oct 17;7(1):263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hansen AR, Stanton TS, Hong MH, Cohen EEW, Mehanna HM, Chisamore MJ, et al. INDUCE-3: A randomized, double-blind study of GSK3359609 (GSK609), an inducible T-cell co-stimulatory (ICOS) agonist antibody, plus pembrolizumab (PE) versus placebo (PL) plus PE for first-line treatment of PD-L1-positive recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). JCO. 2020. May 20;38(15_suppl):TPS6591–TPS6591. [Google Scholar]
- 58.Chakraborty R, Darido C, Liu F, Maselko M, Ranganathan S. Head and Neck Cancer Immunotherapy: Molecular Biological Aspects of Preclinical and Clinical Research. Cancers (Basel). 2023. Jan 30;15(3):852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ghosh S, Shah PA, Johnson FM. Novel Systemic Treatment Modalities Including Immunotherapy and Molecular Targeted Therapy for Recurrent and Metastatic Head and Neck Squamous Cell Carcinoma. Int J Mol Sci. 2022. Jul 17;23(14):7889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vallianou NG, Evangelopoulos A, Kounatidis D, Panagopoulos F, Geladari E, Karampela I, et al. Immunotherapy in Head and Neck Cancer: Where Do We Stand? Curr Oncol Rep. 2023. Aug;25(8):897–912. [DOI] [PubMed] [Google Scholar]
- 61.Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017. Jul 28;357(6349):409–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019. Feb;51(2):202–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang L, Li B, Peng Y, Wu F, Li Q, Lin Z, et al. The prognostic value of TMB and the relationship between TMB and immune infiltration in head and neck squamous cell carcinoma: A gene expression-based study. Oral Oncol. 2020. Nov;110:104943. [DOI] [PubMed] [Google Scholar]
- 64.Valero C, Lee M, Hoen D, Weiss K, Kelly DW, Adusumilli PS, et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021. Feb 1;12(1):729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Behjati S, Tarpey PS. What is next generation sequencing? Arch Dis Child Educ Pract Ed. 2013. Dec;98(6):236–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ma W, Brodie S, Agersborg S, Funari VA, Albitar M. Significant Improvement in Detecting BRAF, KRAS, and EGFR Mutations Using Next-Generation Sequencing as Compared with FDA-Cleared Kits. Mol Diagn Ther. 2017. Oct;21(5):571–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Milbury CA, Creeden J, Yip WK, Smith DL, Pattani V, Maxwell K, et al. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors. PLoS One. 2022;17(3):e0264138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Solis RN, Silverman DA, Birkeland AC. Current Trends in Precision Medicine and Next-Generation Sequencing in Head and Neck Cancer. Curr Treat Options Oncol. 2022. Feb;23(2):254–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Freedman AN, Klabunde CN, Wiant K, Enewold L, Gray SW, Filipski KK, et al. Use of Next-Generation Sequencing Tests to Guide Cancer Treatment: Results From a Nationally Representative Survey of Oncologists in the United States. JCO Precis Oncol. 2018. Nov;2:PO.18.00169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang Y, Springer S, Mulvey CL, Silliman N, Schaefer J, Sausen M, et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci Transl Med. 2015. Jun 24;7(293):293ra104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Economopoulou P, Spathis A, Kotsantis I, Maratou E, Anastasiou M, Moutafi MK, et al. Next-generation sequencing (NGS) profiling of matched tumor and circulating tumor DNA (ctDNA) in head and neck squamous cell carcinoma (HNSCC). Oral Oncol. 2023. Apr;139:106358. [DOI] [PubMed] [Google Scholar]
- 72.Lango MN. Circulating Human Papillomavirus Tumor DNA-Ready for Prime Time? JAMA Otolaryngol Head Neck Surg. 2023. Jul 9; [DOI] [PubMed] [Google Scholar]
- 73.Ferrandino RM, Chen S, Kappauf C, Barlow J, Gold BS, Berger MH, et al. Performance of Liquid Biopsy for Diagnosis and Surveillance of Human Papillomavirus-Associated Oropharyngeal Cancer. JAMA Otolaryngol Head Neck Surg. 2023. Jul 9;e231937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.S C, C V, M L, L LP, G F, C R, et al. A multicenter randomized trial for quality of life evaluation by non-invasive intelligent tools during post-curative treatment follow-up for head and neck cancer: Clinical study protocol. Frontiers in oncology [Internet]. 2023. Jan 31 [cited 2023 Sep 26];13. Available from: https://pubmed.ncbi.nlm.nih.gov/36798825/ [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wang X, Li BB. Deep Learning in Head and Neck Tumor Multiomics Diagnosis and Analysis: Review of the Literature. Front Genet. 2021;12:624820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dhillon K, Manji J, Tapia Céspedes M, Prasad J, Kleid S, Flatman S, et al. Use of telemedicine consultations in head and neck cancer: patient perceptions, acceptability and accessibility. ANZ J Surg. 2022. Jun;92(6):1415–22. [DOI] [PubMed] [Google Scholar]
- 77.Alemi AS, Seth R, Heaton C, Wang SJ, Knott PD. Comparison of Video and In-person Free Flap Assessment following Head and Neck Free Tissue Transfer. Otolaryngol Head Neck Surg. 2017. Jun;156(6):1035–40. [DOI] [PubMed] [Google Scholar]
- 78.Forner D, Noel CW, Shuman AG, Hong P, Corsten M, Rac VE, et al. Shared Decision-making in Head and Neck Surgery: A Review. JAMA Otolaryngol Head Neck Surg. 2020. Sep 1;146(9):839–44. [DOI] [PubMed] [Google Scholar]
- 79.van de Weerd C, Ebbers T, Smilde DEM, van Tol-Geerdink JJ, Takes RP, van den Broek GB, et al. Evaluation of a remote monitoring app in head and neck cancer follow-up care. Cancer Med. 2023. Jul;12(14):15552–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Love RL, Garland R, Ronald M, Alley P, Mutu-Grigg J, Aramoana-Arlidge J, et al. Cultural competency in otolaryngology-head and neck surgery training in Aotearoa, New Zealand. ANZ J Surg. 2020. Dec;90(12):2393–5. [DOI] [PubMed] [Google Scholar]
- 81.Truesdale CM, Baugh RF, Brenner MJ, Loyo M, Megwalu UC, Moore CE, et al. Prioritizing Diversity in Otolaryngology-Head and Neck Surgery: Starting a Conversation. Otolaryngol Head Neck Surg. 2021. Feb;164(2):229–33. [DOI] [PubMed] [Google Scholar]