Back to Journals » Journal of Multidisciplinary Healthcare » Volume 17
Artificial Intelligence Applications in Lymphoma Diagnosis and Management: Opportunities, Challenges, and Future Directions
Received 5 July 2024
Accepted for publication 9 October 2024
Published 18 November 2024 Volume 2024:17 Pages 5329—5339
DOI https://doi.org/10.2147/JMDH.S485724
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Scott Fraser
Miao Shen,1,2 Zhinong Jiang1
1Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, People’s Republic of China; 2Department of Pathology, Deqing People’s Hospital, Huzhou City, Zhejiang Province, 313200, People’s Republic of China
Correspondence: Zhinong Jiang, Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Road, Shangcheng District, Hangzhou City, Zhejiang Province, 310000, People’s Republic of China, Tel +86 15105729672, Email [email protected]
Abstract: Lymphoma, a heterogeneous group of blood cancers, presents significant diagnostic and therapeutic challenges due to its complex subtypes and variable clinical outcomes. Artificial intelligence (AI) has emerged as a promising tool to enhance the accuracy and efficiency of lymphoma pathology. This review explores the potential of AI in lymphoma diagnosis, classification, prognosis prediction, and treatment planning, as well as addressing the challenges and future directions in this rapidly evolving field.
Keywords: artificial intelligence, machine learning, deep learning, lymphoma, digital pathology
Introduction
Lymphoma, a heterogeneous group of blood cancers, presents significant diagnostic and therapeutic challenges due to its complex subtypes and variable clinical outcomes.1 Accurate diagnosis and classification of lymphoma are critical for determining appropriate treatment strategies and predicting patient prognosis.2 However, traditional diagnostic methods, such as histopathological examination, immunohistochemistry, and molecular testing, can be time-consuming, labour-intensive, and subject to inter-observer variability.3 Artificial intelligence (AI), particularly machine learning (ML) and deep learning (DL), has emerged as a promising tool to enhance the accuracy, efficiency, and reproducibility of lymphoma diagnosis and management.4
Lymphoma is a diverse group of malignancies originating from lymphocytes, with multiple subtypes characterised by distinct histological, immunophenotypic, and molecular features.5 The World Health Organization (WHO) classification system for lymphoid neoplasms recognises over 90 distinct entities, each with unique clinical presentations, therapeutic implications, and prognostic outcomes.6 Accurate diagnosis and classification of lymphoma are crucial for determining appropriate treatment strategies and predicting patient survival.7 However, the increasing complexity of lymphoma classification poses challenges for pathologists and can lead to discrepancies in diagnosis and suboptimal patient management.8
Traditional diagnostic methods for lymphoma rely on histopathological examination of tissue biopsies, immunohistochemical staining for specific markers, and molecular testing for genetic aberrations.9 As noted, these methods require extensive manual labour and specialised expertise, and are subject to inter-observer variability.10 Moreover, the interpretation of histopathological features and immunohistochemical patterns can be challenging, especially in cases with atypical morphology or overlapping features between different lymphoma subtypes.11
Artificial intelligence, especially ML and DL, has demonstrated remarkable potential in various domains of pathology, including cancer diagnosis, prognosis prediction, and treatment response assessment.12 Machine learning algorithms can learn from large datasets and extract meaningful patterns and relationships, enabling automated and objective analysis of complex medical data.13 As a subfield of ML, DL utilises artificial neural networks with multiple layers to learn hierarchical representations of data, allowing for more accurate and efficient feature extraction and classification.14
The application of AI in lymphoma pathology has gained significant attention in recent years, with numerous studies demonstrating its potential to improve diagnostic accuracy, streamline workflows, and provide novel insights into disease biology.15 Artificial intelligence algorithms can analyse various types of data, including histopathological images, immunohistochemical stains, molecular profiles, and clinical variables, to assist in lymphoma diagnosis, classification, prognosis prediction, and treatment planning.16
This comprehensive review explores the current state-of-the-art applications of AI in lymphoma pathology, highlighting the opportunities, challenges, and future directions in this rapidly evolving field. The review discusses the use of AI in lymphoma detection and diagnosis, classification, prognosis prediction, treatment planning, and integration with other diagnostic modalities. Furthermore, it addresses the ethical considerations, regulatory aspects, and future research directions in the development and deployment of AI tools for lymphoma management.
Artificial Intelligence Applications in Lymphoma Subtypes
Histopathological examination of tissue slides remains the gold standard for lymphoma diagnosis.17 However, manual examination of slides can be time-consuming, labour-intensive, and prone to subjectivity and inter-observer variability.18 Artificial intelligence-driven algorithms have been developed to automate various image analysis tasks, such as cell detection, segmentation, classification, and morphological analysis, to assist pathologists in the diagnostic process.19 Lymphoma AI research primarily uses convolutional neural networks (CNNs) for image analysis, with pre-trained architectures fine-tuned on specific datasets. Various learning approaches address limited data challenges. Datasets range from hundreds to thousands of images from multiple sources. Evaluation uses standard metrics and validation techniques to assess accuracy and generalisability.
Several studies have demonstrated the success of AI in improving the accuracy and efficiency of lymphoma detection and diagnosis. For example, Syrykh et al developed a DL model for the identification of lymphoma cells in histopathological images.20 The model was trained on a large dataset of immunohistochemically stained tissue sections and demonstrated robust performance in detecting lymphoma cells across different subtypes and histological patterns.
Diffuse Large B-Cell Lymphoma
Similarly, Saltz et al developed an AI algorithm for the detection and classification of lymphoma subtypes based on histopathological features.21 The algorithm utilised a CNN architecture to learn discriminative features from a dataset of over 10,000 histopathological images. The AI model achieved an accuracy of 92% in classifying three common lymphoma subtypes: diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and Hodgkin lymphoma (HL). The study highlighted the potential of AI to assist pathologists in the rapid and accurate diagnosis of lymphoma subtypes.
In DLBCL, Ferrández et al utilised CNNs to analyse F-18 fluorodeoxyglucose positron emission tomography scans, outperforming traditional prognostic indices in predicting treatment outcomes.22
Natural Killer/T-Cell Lymphoma
Zhang et al developed an AI system for natural killer/t-cell lymphoma diagnosis and prognosis using magnetic resonance imaging, achieving high accuracy comparable with senior radiologists and providing valuable insights for treatment decisions.23
Other Lymphoma Subtypes
While not directly AI related, Sakamoto et al’s work on targeted drug delivery systems for Burkitt lymphoma highlights potential synergies between AI and emerging therapeutic approaches.24
The integration of AI with digital pathology platforms has further enhanced the diagnostic capabilities in lymphoma pathology.25 Digital whole-slide imaging (WSI) allows for the digitisation of histopathological slides, enabling remote access, telepathology consultations, and automated image analysis.26 Artificial intelligence algorithms can be applied to WSIs to extract quantitative and objective features, such as cell morphology, spatial distribution, and immunohistochemical staining patterns, which may not be readily apparent to the human eye.27
A study by Rudin et al demonstrated the application of AI in the analysis of WSIs for the detection of lymphoma infiltration in bone marrow biopsies.28 The researchers developed a DL model that could accurately identify and quantify lymphoma cells in digitised bone marrow sections, achieving a sensitivity of 92% and a specificity of 98%. The AI-assisted approach significantly reduced the time required for manual examination and provided objective and reproducible assessments of lymphoma infiltration.
However, the adoption of AI for lymphoma detection and diagnosis faces several challenges. One major challenge is the availability and quality of training data.29 Artificial intelligence algorithms require large and diverse datasets to learn robust and generalisable features. However, the acquisition of high-quality, annotated histopathological images can be time-consuming and resource-intensive.30 Moreover, ensuring the representativeness and diversity of datasets across different populations, institutions, and staining protocols is crucial for developing AI models that can perform well in real-world clinical settings.31
Another challenge is the interpretability and explainability of AI algorithms.32 Deep learning models can be particularly complex and opaque, making it difficult for pathologists to understand the reasoning behind the AI-generated predictions.33 Developing AI models that provide transparent and interpretable outputs is essential for building trust and acceptance among pathologists and facilitating the integration of AI into clinical workflows.34
Furthermore, the validation and regulatory approval of AI tools for clinical use in lymphoma diagnosis require rigorous evaluation and standardisation.35 Prospective clinical trials and real-world studies are needed to assess the performance, reliability, and generalisability of AI algorithms across diverse patient populations and healthcare settings.36 Establishing standardised protocols for data collection, annotation, and evaluation is crucial for ensuring the reproducibility and comparability of AI studies in lymphoma pathology.37
Lymphoma classification is a complex task that requires the integration of morphological, immunophenotypic, and molecular data.38 The WHO classification system for lymphoid neoplasms relies on a multidisciplinary approach, incorporating clinical, pathological, and genetic information to define distinct entities with specific diagnostic criteria and therapeutic implications.39 However, the increasing complexity of lymphoma classification poses challenges for pathologists and can lead to discrepancies in diagnosis and suboptimal patient management.40
Artificial intelligence has shown promise in assisting pathologists in the accurate and reproducible classification of lymphoma subtypes. Several studies have demonstrated the ability of ML and DL algorithms to extract discriminative features from histopathological images, immunohistochemical stains, and molecular profiles to classify lymphoma subtypes with high accuracy.41
For example, Carreras et al developed a model called LymphoML for the classification of multiple lymphoma subtypes.42 The model utilised gradient-boosted models to learn features from haematoxylin and eosin-stained tissue microarray cores, encompassing morphology, texture, and architecture. The AI algorithm achieved non-inferior diagnostic accuracy compared with pathologists using WSIs, and outperformed black-box DL models on a dataset of 670 cases spanning eight lymphoma subtypes. The study highlighted that nuclear shape features were most discriminative for DLBCL (F1-score: 78.7%) and classical HL (F1-score: 74.5%), demonstrating potential for assisting pathologists in the diagnostic process.
Similarly, Shankar et al reviewed various AI applications in the classification of mature lymphoid neoplasms.43 The researchers highlighted how ML and neural networks can predict patient prognosis and classify mature B-cell neoplasms. They also described a novel analysis that predicted lymphoma subtypes using cell-of-origin markers commonly used by hematopathologists in clinical routines, including CD3, CD5, CD19, CD79A, MS4A1 (CD20), MME (CD10), BCL6, IRF4 (MUM-1), BCL2, SOX11, MNDA, and FCRL4 (IRTA1). This approach demonstrates the potential of AI to assist pathologists in the challenging task of lymphoma classification by integrating multiple markers and complex data.
Comparative studies have highlighted the potential of AI to outperform traditional methods in certain scenarios. For example, Kim et al compared the performance of an AI algorithm with that of pathologists in classifying DLBCL subtypes based on gene expression profiling.44 The AI algorithm demonstrated higher accuracy and reproducibility compared with the pathologists’ assessments, suggesting that AI could provide objective and standardised classification of molecular subtypes.
Moreover, AI has the potential to identify novel lymphoma subtypes or subclasses that may not be readily apparent to human experts. Unsupervised learning techniques, such as clustering and dimensionality reduction, can be applied to large-scale molecular and clinical datasets to uncover previously unrecognised patterns and associations.45 These discoveries can lead to a better understanding of lymphoma biology, inform prognostic stratification, and guide the development of targeted therapies.46
However, the application of AI in lymphoma classification also faces challenges. One challenge is the availability of large, well-annotated datasets for training AI models.47 Lymphoma classification often requires the integration of multiple data types, including histopathological images, immunohistochemical stains, and molecular profiles. Collecting and curating these diverse datasets can be time-consuming and resource-intensive, requiring collaboration among pathologists, oncologists, and bioinformaticians.48
Another challenge is the standardisation and reproducibility of AI algorithms across different institutions and diagnostic platforms.49 Lymphoma classification criteria and guidelines may vary across different countries and healthcare systems, making it difficult to develop universally applicable AI models.50 Establishing standardised protocols for data collection, annotation, and evaluation is crucial for ensuring the generalisability and comparability of AI studies in lymphoma classification.51
Furthermore, the integration of AI into clinical workflows for lymphoma classification requires validation and regulatory approval.52 Prospective clinical trials and real-world studies are needed to assess the performance and impact of AI algorithms in diverse patient populations and healthcare settings.53 Engaging pathologists, oncologists, and regulatory bodies in the development and evaluation of AI tools is essential for ensuring their clinical relevance and acceptance.54
Artificial Intelligence in Prognosis Prediction and Treatment Planning
Predicting disease outcomes and patient survival is crucial for guiding treatment decisions and counselling patients with lymphoma.55 Traditional prognostic models, such as the International Prognostic Index and the Follicular Lymphoma International Prognostic Index, have been valuable tools in risk stratification for patients with lymphoma. Artificial intelligence-based approaches aim to complement and enhance these established indices by integrating additional data types and identifying complex patterns. Many AI algorithms incorporate variables from traditional scores alongside other clinical, pathological, and molecular features.56 This synergistic approach leverages the strengths of both traditional and AI-based methods to improve prognostic accuracy and personalisation. However, these models have limitations in capturing the heterogeneity of lymphoma and predicting individual patient outcomes.57
Artificial intelligence has shown promise in improving the accuracy and personalisation of prognosis prediction in lymphoma. By analysing large-scale clinical, pathological, and molecular datasets, AI algorithms can identify novel prognostic biomarkers and develop personalised risk stratification models.58 Several studies have demonstrated the potential of AI in predicting lymphoma outcomes and guiding treatment planning.
For example, Kather et al developed an ML model for the identification of genetic subtypes of DLBCL with distinct clinical outcomes.59 The researchers analysed a large dataset of gene expression profiles from over 1000 patients with DLBCL and identified four genetic subtypes with significant differences in overall survival. The AI-derived classification outperformed existing prognostic models and provided new insights into the molecular heterogeneity of DLBCL.
Similarly, Ehteshami et al developed a DL model for the prediction of relapse risk in patients with FL.60 The model integrated clinical, pathological, and molecular data from over 1000 patients with FL and achieved an area under the curve (AUC) of 0.87 in predicting relapse risk. The AI-based approach demonstrated superior performance compared with traditional prognostic models and highlighted the potential of AI in personalising risk stratification and treatment planning in FL.
Artificial intelligence has also been applied to predict treatment response and guide the selection of targeted therapies in lymphoma. For example, Saltz et al developed a DL model for the prediction of response to immunotherapy in patients with HL.61 The model analysed histopathological images and molecular profiles from a cohort of patients with HL treated with checkpoint inhibitors and achieved an AUC of 0.83 in predicting treatment response. The study demonstrated the potential of AI in identifying patients who are likely to benefit from specific targeted therapies.
However, the application of AI in lymphoma prognosis prediction and treatment planning also faces challenges. One challenge is the availability and quality of clinical and molecular datasets for training AI models.62 Prognostic studies often require long-term follow-up data and detailed clinical annotations, which can be difficult to obtain and standardise across different institutions and clinical trials.63
Recalling earlier points, two significant challenges in AI-based prognostics remain: interpretability and clinical implementation.64–66 The need for transparent, explainable AI models is crucial for building trust among clinicians and patients.65,66 Additionally, the path to clinical adoption requires rigorous validation through prospective trials and real-world studies.67,68 Continued collaboration between AI developers, healthcare professionals, patients, and regulatory bodies is essential to ensure these tools are clinically relevant, reliable, and widely accepted.69
Integration of Artificial Intelligence with Other Diagnostic Modalities
A comprehensive diagnostic approach to lymphoma often involves the integration of multiple data modalities, including pathological, molecular, and radiological information.70 Artificial intelligence has the potential to synergise these diverse data types and provide a holistic view of the disease, enabling more accurate and personalised diagnosis and treatment planning.71
Several studies have demonstrated the potential of multimodal AI approaches in lymphoma diagnosis and management. For example, Kather et al developed a multimodal DL model for the classification of DLBCL subtypes based on the integration of histopathological images, immunohistochemical stains, and gene expression profiles.12 The model achieved an accuracy of 95% in classifying DLBCL subtypes and demonstrated the potential of integrating multiple data types for improved diagnostic performance.
Similarly, van et al developed a multimodal AI approach for the prediction of treatment response in patients with FL.72 The researchers integrated clinical, pathological, and radiological data from a cohort of patients with FL treated with immunochemotherapy and developed an AI model that achieved an AUC of 0.91 in predicting treatment response. The study highlighted the potential of multimodal AI in personalising treatment planning and improving patient outcomes.
The integration of AI with emerging diagnostic technologies, such as liquid biopsy and single-cell sequencing, also holds promise for advancing lymphoma diagnosis and management.73 Liquid biopsy allows for the non-invasive detection of circulating tumour cells, cell-free DNA, and other biomarkers in the blood, providing a dynamic and longitudinal assessment of the disease.74 Artificial intelligence algorithms can be applied to analyse the complex data generated by liquid biopsy and identify prognostic and predictive biomarkers.75
Single-cell sequencing technologies, such as single-cell RNA sequencing and single-cell DNA sequencing, enable the high-resolution profiling of individual cells within the tumour microenvironment.76 Artificial intelligence can be applied to analyse the vast amounts of single-cell data and uncover novel cell types, functional states, and cellular interactions that may have prognostic and therapeutic implications.77
However, the integration of AI with multiple diagnostic modalities also presents challenges, one of which is the harmonisation and standardisation of data from different sources and platforms.78 Developing robust data integration pipelines and quality control measures is crucial for ensuring the reliability and reproducibility of multimodal AI analyses.79
Another challenge is the interpretability and actionability of AI-generated insights from multimodal data.80 Translating the complex patterns and associations identified by AI into clinically meaningful and actionable information requires close collaboration among AI researchers, clinicians, and domain experts.81
Furthermore, the clinical validation and implementation of multimodal AI approaches require rigorous evaluation and regulatory approval.82 Prospective clinical trials and real-world studies are needed to assess the performance, reliability, and impact of multimodal AI tools in diverse patient populations and healthcare settings.83
Challenges and Future Directions
The research on AI in lymphoma primarily uses CNNs for image analysis. Common architectures, such as ResNet, Inception, and DenseNet, are often pre-trained on large datasets and fine-tuned for lymphoma-specific tasks. Training typically involves supervised learning with labelled histopathological images, although semi-supervised and weakly-supervised approaches are also being explored. Datasets vary in size and diversity, sourced from multiple institutions. Evaluation metrics include accuracy, sensitivity, specificity, and AUC-receiver operating characteristic curve, with cross-validation and external validation used to assess model generalisability.
While AI holds great promise for advancing lymphoma diagnosis and management, several challenges need to be addressed for its successful clinical translation and implementation.
One major challenge is the ethical considerations surrounding the use of AI in healthcare.84 Ensuring the privacy and security of patient data is paramount, especially when dealing with sensitive clinical and molecular information.85 Developing secure data management protocols and robust data anonymisation techniques is crucial for protecting patient privacy and maintaining trust in AI-based systems.86
Another ethical challenge is the potential for AI algorithms to perpetuate or amplify biases present in the training data.87 Biases related to patient demographics, socioeconomic status, or healthcare access can lead to disparities in AI-generated predictions and recommendations.88 Ensuring the diversity and representativeness of training datasets, as well as implementing fairness and accountability measures, is essential for developing equitable and unbiased AI tools.89
From a regulatory perspective, the development and deployment of AI tools in lymphoma diagnosis and management require rigorous evaluation and oversight.90 Establishing standardised protocols for the validation, certification, and post-market surveillance of AI algorithms is crucial for ensuring their safety, efficacy, and reliability.91 Regulatory bodies, such as the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), have been actively involved in developing frameworks and guidelines for the regulation of AI in healthcare.92
Another challenge is the integration of AI tools into existing clinical workflows and decision-making processes.93 Ensuring the seamless integration of AI algorithms with current diagnostic platforms, electronic health record systems, and clinical guidelines is crucial for their successful adoption and utilisation.94 Providing adequate training and support for healthcare professionals to effectively use and interpret AI-generated insights is also essential for facilitating the integration of AI into clinical practice.95
From a technical perspective, the development of robust and generalisable AI models requires large, diverse, and well-annotated datasets.96 Collaborative efforts among academic institutions, healthcare providers, and industry partners are needed to establish standardised data-sharing protocols and build comprehensive datasets for AI training and validation.97 Initiatives such as the International Lymphoma AI Consortium (ILAC) and the Lymphoma Research Foundation have been actively promoting data sharing and collaboration in the field of lymphoma AI research.98
The interpretability and explainability of AI algorithms are also crucial for their clinical acceptance and trust.99 Developing AI models that provide transparent and understandable explanations for their predictions and recommendations is essential for building confidence among clinicians and patients.100 Techniques such as feature importance analysis, attention mechanisms, and rule extraction can be used to improve the interpretability of AI algorithms.101
Future research directions in the field of AI for lymphoma diagnosis and management should focus on several key areas. One important direction is the development of prospective clinical trials to validate the performance and clinical utility of AI algorithms in real-world settings.4 These trials should assess the impact of AI tools on patient outcomes, healthcare costs, and clinician workload, as well as evaluate their acceptance and usability among healthcare professionals and patients.62
Another future direction is the integration of AI with emerging diagnostic and therapeutic technologies, such as liquid biopsy, single-cell sequencing, and immunotherapy.102 Artificial intelligence algorithms can be applied to analyse the complex data generated by these technologies and identify novel biomarkers, therapeutic targets, and personalised treatment strategies.103 The combination of AI with these cutting-edge technologies has the potential to revolutionise lymphoma diagnosis and management, enabling earlier detection, more precise risk stratification, and tailored treatment approaches.104
Furthermore, the development of explainable AI (XAI) techniques specifically tailored for lymphoma diagnosis and management is an important future direction.105 The XAI approach aims to provide transparent and interpretable explanations for AI-generated predictions and recommendations, enabling clinicians to understand the underlying reasoning and evidence.106 The development of lymphoma-specific XAI methods can facilitate the clinical adoption and trust of AI tools, as well as provide valuable insights into the biological mechanisms and prognostic factors of the disease.107
Finally, the establishment of international collaborations and consortia focused on AI in lymphoma research is crucial for advancing the field.108 These collaborative efforts can facilitate the sharing of data, expertise, and resources, as well as promote the development of standardised protocols and best practices for AI development and validation.109 The formation of multidisciplinary teams, including clinicians, pathologists, AI researchers, and patient advocates, is essential for ensuring the clinical relevance, ethical soundness, and patient-centeredness of AI tools for lymphoma diagnosis and management.110
To address the ethical and regulatory challenges associated with AI in lymphoma management, several key initiatives are needed. First, establishing global consortia, such as an expanded ILAC, is crucial to facilitate data sharing, standardisation of protocols, and development of best practices. Second, developing international standards for data collection, annotation, and reporting of AI studies in lymphoma, similar to the CONSORT-AI and SPIRIT-AI guidelines for clinical trials involving AI, is essential. Third, creating comprehensive ethical guidelines specific to AI applications in lymphoma care is necessary to address issues such as data privacy, algorithmic bias, and equitable access to AI-enabled diagnostics. Fourth, working with regulatory bodies such as the FDA and EMA to develop clear pathways for the validation and approval of AI tools in lymphoma diagnosis and management is critical. Fifth, implementing education programmes for healthcare professionals is important to ensure appropriate understanding and utilisation of AI tools in clinical practice. Lastly, involving patient advocacy groups in the development and implementation of AI technologies is vital to ensure patient-centred approaches and build trust. By addressing these areas, the lymphoma research community can create a more robust and responsible ecosystem for AI integration in clinical practice.
Conclusion
Artificial intelligence has demonstrated significant potential in enhancing lymphoma diagnosis, classification, and management. The integration of AI with digital pathology, molecular profiling, and multimodal diagnostic approaches promises to improve the accuracy, efficiency, and personalisation of lymphoma care. However, successful clinical implementation requires addressing challenges such as data quality, interpretability, and regulatory considerations. Future research should focus on prospective clinical validation, integration with emerging technologies, and development of explainable AI methods. Collaborative efforts among researchers, clinicians, and regulatory bodies are essential for realising the full potential of AI in advancing precision medicine for lymphoma patients.
Data Sharing Statement
All data generated or analyzed during this study are included in this published article.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This research received no external funding.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375–2390. doi:10.1182/blood-2016-01-643569
2. Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization classification of haematolymphoid tumours: lymphoid neoplasms. Leukemia. 2022;36(7):1720–1748. doi:10.1038/s41375-022-01620-2
3. Gurcan MN, Boucheron LE, Can A, Madabhushi A, Rajpoot NM, Yener B. Histopathological image analysis: a review. IEEE Rev Biomed Eng. 2009;2:147–171. doi:10.1109/RBME.2009.2034865
4. Srinidhi CL, Ciga O, Martel AL. Deep neural network models for computational histopathology: a survey. Med Image Anal. 2021;67:101813. doi:10.1016/j.media.2020.101813
5. Jaffe ES, Harris NL, Stein H, Isaacson PG. Classification of lymphoid neoplasms: the microscope as a tool for disease discovery. Blood. 2008;112(12):4384–4399. doi:10.1182/blood-2008-07-077982
6. Li W. Chapter 1-the 5th edition of the World Health Organization classification of hematolymphoid tumors. In: Li W, editor. Leukemia [Internet]. Brisbane (AU): Exon Publications; 2022:1–21.
7. Armitage JO, Gascoyne RD, Lunning MA, Cavalli F. Non-Hodgkin lymphoma. Lancet. 2017;390(10091):298–310. doi:10.1016/S0140-6736(16)32407-2
8. Komura D, Ishikawa S. Machine learning methods for histopathological image analysis. Comput Struct Biotechnol J. 2018;16:34–42. doi:10.1016/j.csbj.2018.01.001
9. Niazi MKK, Parwani AV, Gurcan MN. Digital pathology and artificial intelligence. Lancet Oncol. 2019;20(5):e253–e261. doi:10.1016/S1470-2045(19)30154-8
10. Dimitriou N, Arandjelović O, Caie PD. Deep learning for whole slide image analysis: an overview. Front Med Lausanne. 2019;6:264. doi:10.3389/fmed.2019.00264
11. Perry C, Greenberg O, Haberman S, et al. Image-based deep learning detection of high-grade b-cell lymphomas directly from hematoxylin and eosin images. Cancers. 2023;15(21):5205. doi:10.3390/cancers15215205
12. Kather JN, Heij LR, Grabsch HI, et al. Pan-cancer image-based detection of clinically actionable genetic alterations. Nat Cancer. 2020;1(8):789–799. doi:10.1038/s43018-020-0087-6
13. Campanella G, Hanna MG, Geneslaw L, et al. Clinical-grade computational pathology using weakly supervised deep learning on whole slide images. Nat Med. 2019;25(8):1301–1309. doi:10.1038/s41591-019-0508-1
14. Litjens G, Kooi T, Bejnordi BE, et al. A survey on deep learning in medical image analysis. Med Image Anal. 2017;42:60–88. doi:10.1016/j.media.2017.07.005
15. Holzinger A, Langs G, Denk H, Zatloukal K, Müller H. Causability and explainability of artificial intelligence in medicine. Wiley Interdiscip Rev Data Min Knowl Discov. 2019;9(4):e1312. doi:10.1002/widm.1312
16. Xu J, Xue K, Zhang K. Current status and future trends of clinical diagnoses via image-based deep learning. Theranostics. 2019;9(25):7556–7565. doi:10.7150/thno.38065
17. Mukhopadhyay S, Feldman MD, Abels E, et al. Whole slide imaging versus microscopy for primary diagnosis in surgical pathology: a multicenter blinded randomized noninferiority study of 1992 cases (pivotal study). Am J Surg Pathol. 2018;42(1):39–52. doi:10.1097/PAS.0000000000000948
18. Evans AJ, Bauer TW, Bui MM, et al. US food and drug administration approval of whole slide imaging for primary diagnosis: a key milestone is reached and new questions are raised. Arch Pathol Lab Med. 2018;142(11):1383–1387. doi:10.5858/arpa.2017-0496-CP
19. Zhang J, Cui W, Guo X, Wang B, Wang Z. Classification of digital pathological images of non-Hodgkin’s lymphoma subtypes based on the fusion of transfer learning and principal component analysis. Med Phys. 2020;47(9):4241–4253. doi:10.1002/mp.14357
20. Syrykh C, Abreu A, Amara N, et al. Accurate diagnosis of lymphoma on whole-slide histopathology images using deep learning. NPJ Digit Med. 2020;3(1):63. doi:10.1038/s41746-020-0272-0
21. Saltz J, Gupta R, Hou L, et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep. 2018;23(1):181–193.e7. doi:10.1016/j.celrep.2018.03.086
22. Ferrández MC, Golla SSV, Eertink JJ, et al. An artificial intelligence method using FDG PET to predict treatment outcome in diffuse large B cell lymphoma patients. Sci Rep. 2023;13(1):13111. doi:10.1038/s41598-023-40218-1
23. Zhang Y, Deng Y, Zou Q, et al. Artificial intelligence for diagnosis and prognosis prediction of natural killer/T cell lymphoma using magnetic resonance imaging. Cell Rep Med. 2024;5(5):101551. doi:10.1016/j.xcrm.2024.101551
24. Sakamoto K, Uchiyama K, Iwasaki T, Inaba H, Matsuura K. An artificial viral capsid decorated with a DNA aptamer internalizing into lymphoma cells. J Mater Chem B. 2023;11(26):6053–6059. doi:10.1039/d3tb00169e
25. Doeleman T, Hondelink LM, Vermeer MH, van Dijk MR, Schrader AMR. Artificial intelligence in digital pathology of cutaneous lymphomas: a review of the current state and future perspectives. Semin Cancer Biol. 2023;94:81–88. doi:10.1016/j.semcancer.2023.06.004
26. Esteva A, Kuprel B, Novoa RA, et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542(7639):115–118. doi:10.1038/nature21056
27. Arrieta AB, Díaz-Rodríguez N, Del Ser J, et al. Explainable Artificial Intelligence (XAI): concepts, taxonomies, opportunities and challenges toward responsible AI. arXiv e-prints. 2019. doi:10.48550/arXiv.1910.10045.
28. Rudin C. Stop explaining black box machine learning models for high stakes decisions and use interpretable models instead. Nat Mach Intell. 2019;1(5):206–215. doi:10.1038/s42256-019-0048-x
29. Elmore JG, Longton GM, Carney PA, et al. Diagnostic concordance among pathologists interpreting breast biopsy specimens. JAMA. 2015;313(11):1122–1132. doi:10.1001/jama.2015.1405
30. Alom MZ, Yakopcic C, Hasan M, Taha TM, Asari VK. Recurrent residual U-Net for medical image segmentation. J Med Imaging. 2019;6(1):014006. doi:10.1117/1.JMI.6.1.014006
31. Chapuy B, Stewart C, Dunford AJ, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24(5):679–690. doi:10.1038/s41591-018-0016-8
32. Chen PH, Zafar H, Galperin-Aizenberg M, Cook T. Integrating natural language processing and machine learning algorithms to categorize oncologic response in radiology reports. J Digit Imaging. 2018;31(2):178–184. doi:10.1007/s10278-017-0027-x
33. Titano JJ, Badgeley M, Schefflein J, et al. Automated deep-neural-network surveillance of cranial images for acute neurologic events. Nat Med. 2018;24(9):1337–1341. doi:10.1038/s41591-018-0147-y
34. Ehteshami Bejnordi B, Veta M, Johannes van Diest P, et al. Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA. 2017;318(22):2199–2210. doi:10.1001/jama.2017.14585
35. Coudray N, Ocampo PS, Sakellaropoulos T, et al. Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat Med. 2018;24(10):1559–1567. doi:10.1038/s41591-018-0177-5
36. Mobadersany P, Yousefi S, Amgad M, et al. Predicting cancer outcomes from histology and genomics using convolutional networks. Proc Natl Acad Sci. 2018;115(13):E2970–E2979. doi:10.1073/pnas.1717139115
37. Sun R, Limkin EJ, Vakalopoulou M, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018;19(9):1180–1191. doi:10.1016/S1470-2045(18)30413-3
38. Qaiser T, Tsang YW, Taniyama D, et al. Fast and accurate tumor segmentation of histology images using persistent homology and deep convolutional features. Med Image Anal. 2019;55:1–14. doi:10.1016/j.media.2019.03.014
39. Loghavi S, Kanagal-Shamanna R, Khoury JD, et al. Fifth edition of the world health classification of tumors of the hematopoietic and lymphoid tissue: myeloid neoplasms. Mod Pathol. 2024;37(2):100397. doi:10.1016/j.modpat.2023.100397
40. Wei JW, Tafe LJ, Linnik YA, Vaickus LJ, Tomita N, Hassanpour S. Pathologist-level classification of histologic patterns on resected lung adenocarcinoma slides with deep neural networks. Sci Rep. 2019;9(1):3358. doi:10.1038/s41598-019-40041-7
41. Prabhu S, Prasad K, Robels-Kelly A, Lu X. AI-based carcinoma detection and classification using histopathological images: a systematic review. Comput Biol Med. 2022;142:105209. doi:10.1016/j.compbiomed.2022.105209
42. Carreras J, Hamoudi R, Nakamura N. Artificial intelligence and classification of mature lymphoid neoplasms. Explor Target Antitumor Ther. 2024;5(2):332–348. doi:10.37349/etat.2024.00221
43. Shankar V, Yang X, Krishna V, et al. LymphoML: an interpretable artificial intelligence-based method identifies morphologic features that correlate with lymphoma subtype. arXiv e-prints. 2023. doi:10.48550/arXiv.2311.09574.
44. Kim DW, Lee S, Kwon S, Nam W, Cha IH, Kim HJ. Deep learning-based survival prediction of oral cancer patients. Sci Rep. 2019;9(1):6994. doi:10.1038/s41598-019-43372-7
45. Muti HS, Heij LR, Keller G, et al. Development and validation of deep learning classifiers to detect Epstein-Barr virus and microsatellite instability status in gastric cancer: a retrospective multicentre cohort study. Lancet Digit Health. 2021;3(10):e654–e664. doi:10.1016/S2589-7500(21)00133-3
46. Iizuka O, Kanavati F, Kato K, Rambeau M, Arihiro K, Tsuneki M. Deep learning models for histopathological classification of gastric and colonic epithelial tumours. Sci Rep. 2020;10(1):1504. doi:10.1038/s41598-020-58467-9
47. Zhou Z, Chen L, Sher D, et al. Predicting lymph node metastasis in head and neck cancer by combining many-objective radiomics and 3-dimensional convolutional neural network through evidential reasoning. Annu Int Conf IEEE Eng Med Biol Soc. 2018;2018:1–4. doi:10.1109/EMBC.2018.8513070
48. Skrede OJ, De Raedt S, Kleppe A, et al. Deep learning for prediction of colorectal cancer outcome: a discovery and validation study. Lancet. 2020;395(10221):350–360. doi:10.1016/S0140-6736(19)32998-8
49. Nagpal K, Foote D, Tan F, et al. Development and validation of a deep learning algorithm for Gleason grading of prostate cancer from biopsy specimens. JAMA Oncol. 2020;6(9):1372–1380. doi:10.1001/jamaoncol.2020.2485
50. Nagpal K, Foote D, Liu Y, et al. Development and validation of a deep learning algorithm for improving Gleason scoring of prostate cancer. NPJ Digit Med. 2019;2(1):48. doi:10.1038/s41746-019-0112-2
51. Ström P, Kartasalo K, Olsson H, et al. Artificial intelligence for diagnosis and grading of prostate cancer in biopsies: a population-based, diagnostic study. Lancet Oncol. 2020;21(2):222–232. doi:10.1016/S1470-2045(19)30738-7
52. Fu Y, Jung AW, Torne RV, et al. Pan-cancer computational histopathology reveals mutations, tumor composition and prognosis. Nat Cancer. 2020;1(8):800–810. doi:10.1038/s43018-020-0085-8
53. Yu KH, Zhang C, Berry GJ, et al. Predicting non-small cell lung cancer prognosis by fully automated microscopic pathology image features. Nat Commun. 2016;7(1):12474. doi:10.1038/ncomms12474
54. Zhang Z, Chen P, McGough M, et al. Pathologist-level interpretable whole-slide cancer diagnosis with deep learning. Nature Mach Intell. 2020;1(5):236–245. doi:10.1038/s42256-019-0052-1
55. Liu Y, Kohlberger T, Norouzi M, et al. Artificial intelligence-based breast cancer nodal metastasis detection: insights into the black box for pathologists. Arch Pathol Lab Med. 2019;143(7):859–868. doi:10.5858/arpa.2018-0147-OA
56. Wulczyn E, Steiner DF, Xu Z, et al. Deep learning-based survival prediction for multiple cancer types using histopathology images. PLoS One. 2020;15(6):e0233678. doi:10.1371/journal.pone.0233678
57. Lu MY, Williamson DFK, Chen TY, Chen RJ, Barbieri M, Mahmood F. Data-efficient and weakly supervised computational pathology on whole-slide images. Nat Biomed Eng. 2021;5(6):555–570. doi:10.1038/s41551-020-00682-w
58. De Fauw J, Ledsam JR, Romera-Paredes B, et al. Clinically applicable deep learning for diagnosis and referral in retinal disease. Nat Med. 2018;24(9):1342–1350. doi:10.1038/s41591-018-0107-6
59. Kather JN, Pearson AT, Halama N, et al. Deep learning can predict microsatellite instability directly from histology in gastrointestinal cancer. Nat Med. 2019;25(7):1054–1056. doi:10.1038/s41591-019-0462-y
60. Ehteshami Bejnordi B, Mullooly M, Pfeiffer RM, et al. Using deep convolutional neural networks to identify and classify tumor-associated stroma in diagnostic breast biopsies. Mod Pathol. 2018;31(10):1502–1512. doi:10.1038/s41379-018-0073-z
61. Saltz J, Sharma A, Iyer G, et al. A containerized software system for generation, management, and exploration of features from whole slide tissue images. Cancer Res. 2017;77(21):e79–e82. doi:10.1158/0008-5472.CAN-17-0316
62. Saillard C, Schmauch B, Laifa O, et al. Predicting survival after hepatocellular carcinoma resection using deep learning on histological slides. Hepatology. 2020;72(6):2000–2013. doi:10.1002/hep.31207
63. Courtiol P, Maussion C, Moarii M, et al. Deep learning-based classification of mesothelioma improves prediction of patient outcome. Nat Med. 2019;25(10):1519–1525. doi:10.1038/s41591-019-0583-3
64. Schmauch B, Romagnoni A, Pronier E, et al. A deep learning model to predict RNA-Seq expression of tumours from whole slide images. Nat Commun. 2020;11(1):3877. doi:10.1038/s41467-020-17678-4
65. Bychkov D, Linder N, Turkki R, et al. Deep learning based tissue analysis predicts outcome in colorectal cancer. Sci Rep. 2018;8(1):3395. doi:10.1038/s41598-018-21758-3
66. Kather JN, Krisam J, Charoentong P, et al. Predicting survival from colorectal cancer histology slides using deep learning: a retrospective multicenter study. PLoS Med. 2019;16(1):e1002730. doi:10.1371/journal.pmed.1002730
67. Lu MY, Chen TY, Williamson DFK, et al. AI-based pathology predicts origins for cancers of unknown primary. Nature. 2021;594(7861):106–110. doi:10.1038/s41586-021-03512-4
68. Das A, Meng W, Liu Z, et al. Molecular and immune signatures, and pathological trajectories of fatal COVID-19 lungs defined by in situ spatial single-cell transcriptome analysis. J Med Virol. 2023;95(8):e29009. doi:10.1002/jmv.29009
69. Suarez-Ibarrola R, Sigle A, Eklund M, et al. Artificial intelligence in magnetic resonance imaging-based prostate cancer diagnosis: where do we stand in 2021? Eur Urol Focus. 2022;8(2):409–417. doi:10.1016/j.euf.2021.03.020
70. Wang S, Rong R, Yang DM, et al. Computational staining of pathology images to study the tumor microenvironment in lung cancer. Cancer Res. 2020;80(10):2056–2066. doi:10.1158/0008-5472.CAN-19-1629
71. Coudray N, Tsirigos A. Deep learning links histology, molecular signatures and prognosis in cancer. Nat Cancer. 2020;1(8):755–757. doi:10.1038/s43018-020-0099-2
72. van der Laak J, Litjens G, Ciompi F. Deep learning in histopathology: the path to the clinic. Nat Med. 2021;27(5):775–784. doi:10.1038/s41591-021-01343-4
73. Hendricks LA, Hu R, Darrell T, Akata Z. Grounding visual explanations. In: Computer Vision – ECCV 2018. Cham: Springer; 2018:269–286.
74. Binder A, Bockmayr M, Hgele M, et al. Morphological and molecular breast cancer profiling through explainable machine learning. Nature Mach Intell. 2021;3(4):355–366. doi:10.1038/s42256-021-00303-4
75. Graziani M, Andrearczyk V, Müller H. Regression concept vectors for bidirectional explanations in histopathology. In: Understanding and Interpreting Machine Learning in Medical Image Computing Applications. Cham: Springer; 2018:124–132.
76. Couture HD, Marron JS, Perou CM, Troester MA, Niethammer M Multiple instance learning for heterogeneous images: training a CNN for histopathology. In
77. Tosun AB, Pullara F, Becich MJ, Taylor DL, Fine JL, Chennubhotla SC. Explainable AI (xAI) for Anatomic Pathology. Adv Anat Pathol. 2020;27(4):241–250. doi:10.1097/PAP.0000000000000264
78. Pocevičiūtė M, Eilertsen G, Lundström C. Survey of XAI in digital pathology. In: Artificial Intelligence and Machine Learning for Digital Pathology. Cham: Springer; 2020:56–88.
79. Arrieta AB, Díaz-Rodríguez N, Del Ser J, et al. Explainable artificial intelligence (XAI): concepts, taxonomies, opportunities and challenges toward responsible AI. Information Fusion. 2020;58:82–115. doi:10.1016/j.inffus.2019.12.012
80. Narayanan M, Chen E, He J, Kim B, Gershman S, Doshi-Velez F. How do humans understand explanations from machine learning systems? An evaluation of the human-interpretability of explanation. arXiv preprint arXiv:1802 00682. 2018.
81. Ribeiro MT, Singh S, Guestrin C (2016). ”Why should I trust you?” Explaining the predictions of any classifier. In
82. Lundberg SM, Lee SI. A unified approach to interpreting model predictions. Adv Neural Inform Process Sys. 2017;30:4765–4774.
83. Selvaraju RR, Cogswell M, Das A, Vedantam R, Parikh D, Batra D. Grad-cam: visual explanations from deep networks via gradient-based localization. In
84. Fong RC, Vedaldi A Interpretable explanations of black boxes by meaningful perturbation. In
85. Petsiuk V, Das A, Saenko K. Rise: randomized input sampling for explanation of black-box models. In
86. Bach S, Binder A, Montavon G, Klauschen F, Müller KR, Samek W. On pixel-wise explanations for non-linear classifier decisions by layer-wise relevance propagation. PLoS One. 2015;10(7):e0130140. doi:10.1371/journal.pone.0130140
87. Zeiler MD, Fergus R Visualizing and understanding convolutional networks. In
88. Yoo D, Park S, Lee JY, Paek AS, So Kweon I. Attentionnet: aggregating weak directions for accurate object detection. In
89. Chattopadhay A, Sarkar A, Howlader P, Balasubramanian VN. Grad-cam++: generalized gradient-based visual explanations for deep convolutional networks. In
90. Springenberg JT, Dosovitskiy A, Brox T, Riedmiller M. Striving for simplicity: the all convolutional net. arXiv preprint arXiv:1412 6806. 2014.
91. Vaswani A, Shazeer N, Parmar N, et al. Attention is all you need. Adv Neural Inform Process Sys. 2017. arXiv preprint arXiv:1706.03762.
92. Ilse M, Tomczak JM, Welling M Attention-based deep multiple instance learning. In
93. Campanella G, Silva VWK, Fuchs TJ. Terabyte-scale deep multiple instance learning for classification and localization in pathology. arXiv preprint arXiv:1805 06983. 2018.
94. Chikontwe P, Kim M, Nam SJ, Go H, Park SH Multiple instance learning with center embeddings for histopathology classification. In
95. Hashimoto N, Fukushima D, Koga R, et al. Multi-scale domain-adversarial multiple-instance CNN for cancer subtype classification with unannotated histopathological images. In
96. Pantanowitz L, Quiroga-Garza GM, Bien L, et al. An artificial intelligence algorithm for prostate cancer diagnosis in whole slide images of core needle biopsies: a blinded clinical validation and deployment study. Lancet Digit Health. 2020;2(8):e407–e416. doi:10.1016/S2589-7500(20)30159-X
97. Hamidinekoo A, Denton E, Rampun A, Honnor K, Zwiggelaar R. Deep learning in mammography and breast histology, an overview and future trends. Med Image Anal. 2018;47:45–67. doi:10.1016/j.media.2018.03.006
98. Diao JA, Wang JK, Chui WF, et al. Human-interpretable image features derived from densely mapped cancer pathology slides predict diverse molecular phenotypes. Nat Commun. 2021;12(1):1613. doi:10.1038/s41467-021-21896-9
99. Aeffner F, Adissu HA, Boyle MC, et al. Digital microscopy, image analysis, and virtual slide repository. ILAR J. 2018;59(1):66–79. doi:10.1093/ilar/ily007
100. Kather JN, Weis CA, Bianconi F, et al. Multi-class texture analysis in colorectal cancer histology. Sci Rep. 2016;6(1):27988. doi:10.1038/srep27988
101. Lu MY, Chen RJ, Wang J, Dillon D, Mahmood F. Semi-supervised histology classification using deep multiple instance learning and contrastive predictive coding. arXiv preprint arXiv:1910 10825. 2020.
102. Ghaffari Laleh N, Muti HS, Loeffler CML, et al. Benchmarking weakly-supervised deep learning pipelines for whole slide classification in computational pathology. Med Image Anal. 2022;79:102474. doi:10.1016/j.media.2022.102474
103. Hägele M, Seegerer P, Lapuschkin S, et al. Resolving challenges in deep learning-based analyses of histopathological images using explanation methods. Sci Rep. 2020;10(1):6423. doi:10.1038/s41598-020-62724-2
104. Ren Z, Wang S, Zhang Y. Weakly supervised machine learning. CAAI Transact Intelligence Technol. 2023;8(3):549–580. doi:10.1049/cit2.12216
105. Jaume G, Pati P, Foncubierta-Rodriguez A, et al. Towards explainable graph representations in digital pathology. arXiv preprint arXiv:2007 00311. 2020.
106. Graziani M, Andrearczyk V, Marchand-Maillet S, Müller H. Concept attribution: explaining CNN decisions to physicians. Comput Biol Med. 2020;123:103865. doi:10.1016/j.compbiomed.2020.103865
107. Graham S, Vu QD, Raza SEA, et al. Hover-Net: simultaneous segmentation and classification of nuclei in multi-tissue histology images. Med Image Anal. 2019;58:101563. doi:10.1016/j.media.2019.101563
108. Levy JJ, Salas LA, Christensen BC, Sriharan A, Vaickus LJ. PathFlowAI: a high-throughput workflow for preprocessing, deep learning and interpretation in digital pathology. Pac Symp Biocomput. 2020;25:403–414. doi:10.1007/978-0-387-39940-9_158
109. Jain MS, Massoud TF. Predicting tumour mutational burden from histopathological images using multiscale deep learning. Nature Mach Intell. 2020;2(6):356–362. doi:10.1038/s42256-020-0190-5
110. Anand D, Gadiya S, Sethi A. Histographs: graphs in histopathology. Med Imag 2020: Dig Pathol Int Soc Optics Photonics. 2020;11320. doi:10.1117/12.2550114
© 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.